Download Clinical and laboratory markers of disease activity and response to

Document related concepts

Gene therapy wikipedia , lookup

Syndemic wikipedia , lookup

Epidemiology wikipedia , lookup

Public health genomics wikipedia , lookup

Disease wikipedia , lookup

Gene therapy of the human retina wikipedia , lookup

Hygiene hypothesis wikipedia , lookup

Alzheimer's disease research wikipedia , lookup

Sjögren syndrome wikipedia , lookup

Multiple sclerosis research wikipedia , lookup

Transcript
Clinical and laboratory markers of disease activity and response to
therapy in South Africans with Rheumatoid Arthritis with disease duration
less than two years
by
Mahmood Moosa Tar Mahomed Ally
in
The Department of Internal Medicine
Faculty of Health Sciences
University of Pretoria
2015
1
DECLARATION
I, Prof Mahmood MTM Ally , hereby declare that the work on which this research is
based is original (except where acknowledgements indicate otherwise) and that
neither the whole work nor part of it, is being or is to be submitted for another degree
at this or any other university.
12 June 2015
-----------------------------------.
Signature
--------------------------Date
2
ACKNOWLEDGEMENTS
“To start something good is your job and to see it completed is the work of your
creator”.
The essence of this GREAT (Gauteng Rheumatoid Evaluation Assessment Trial)
research adventure is to expand our knowledge so as to open the doors of good
health for our patients. A journey which would not have been possible had it not been
for the support, guidance and encouragement of many people.
I would like to acknowledge:
Our patients who despite physical and psycho-social hardships, participated
unconditionally.
Prof Ronald Anderson and Prof Mohamed Tikly for their enduring and exceptional
leadership in steering this project
My co-authors Pieter Meyer, Bridget Hodkinson, and Eustatius Musenge for their
invaluable hard work, patience and dedication.
Prof Gregory Tintinger my supervisor, for his academic guidance.
Prof Fathima Suleman, my sister in law for her academic input.
To my colleagues and supporting staff in the rheumatology units SBAH and CHBH for
their assistance in collecting the data and facilitating preparation of the dissertation.
Staff from the Department of Immunology/ National Health Laboratory Service
University of Pretoria for going the extra mile.
Hannatjie van wyk for assistance with preparation of manuscripts and this dissertation
Ina steyn for secretarial assistance.
Rheumatology Development Fund University of Pretoria, the Connective Tissue
Disease Fund of the University of the Witwatersrand, the Medical Research Council
and the National Health Laboratory Service Research Trust for sponsoring the
project.
My parents for their encouragement and inspiration.
My wife Farhana and children Raihaan, Bilal and Fathima for their forbearance and
understanding support.
3
PUBLICATIONS
Original publications in ISI accredited journals:
1. Ally MM, Hodkinson B, Meyer PW, Musenge E, Tikly M, Anderson R. Serum
matrix metalloproteinase-3 in comparison with acute phase proteins as a
marker of disease activity and radiographic damage in early rheumatoid
arthritis. Mediators of Inflammation 2013; 2013:183653.
2. Hodkinson B, Meyer PWA, Musenge E, Ally MM, Anderson R, Tikly M.
Functional disability and health-related quality of life in South Africans with
early rheumatoid arthritis. Scandinavian Journal of Rheumatology 2012;
41(5):366-74.
3. Hodkinson B, Meyer PW, Musenge E, Ally M, Anderson R, Tikly M.
Exaggerated circulating Th-1 cytokine response in early rheumatoid arthritis
patients with nodules. Cytokine 2012; 60(2):561-4.
4. Hodkinson B, Musenge E, Ally M, Meyer PWA, Anderson R, Tikly M.
Response to traditional disease modifying anti-rheumatic drugs in indigent
South Africans with early rheumatoid arthritis. Clinical Rheumatology 2012;
31(4): 613-619.
5. Meyer PW, Hodkinson B, Ally M, Musenge E, Wadee AA, Fickl H, Tikly M,
Anderson R. HLA-DRB1 shared epitope genotyping using the revised
classification and its association with circulating autoantibodies, acute phase
reactants cytokines and clinical indices of disease activity in a cohort of South
African rheumatoid arthritis patients. Arthritis Research and Therapy 2011;
13(5): R160.
6. Hodkinson B, Meyer PW, Musenge E, Ally MM, Wadee AA, Anderson R, Tikly
M. The diagnostic utility of the anti-CCP antibody test is no better than
rheumatoid factor in South Africans with earl rheumatoid arthritis. Clinical
Rheumatology 2010; 29(6): 615-618.
7. Meyer PWA, Hodkinson B, Ally M, Musenge E, Wadee AA, Fickl H, Tikly M,
Anderson R. Circulating cytokine profiles and their relationships with
autoantibodies, acute phase reactants, and disease activity in patients with
rheumatoid arthritis. Mediators of Inflammation 2010; 2010: 158514.
4
Review articles in ISI accredited journals:
1. Ally, Mahmood MTM. - Biomarkers in rheumatoid arthritis - the old and
new. SA Orthopaedic Journal 2013, vol.12, n.4, pp. 19-22. ISSN 1681-150X.
2. Ally M, Hodkinson B. Rheumatoid Arthritis. South African Family Practice
2014; 56(3): 166-171. doi:10.1080/20786204.2014.932547.
3. Ally MMTM, Visser CC. Rheumatoid arthritis Part 1: Review of the
pathogenesis, clinical features, utility of special investigations and measures of
disease activity in patients with rheumatoid artritis. SA Orthopaedic Journal
2010; vol. 9, no.1, pp. 18-23.
4. Ally MMTM, Visser CC. Rheumatoid arthritis Part II: Review of the
management and surgical considerations of disease-modifying anti-rheumatic
drugs in rheumatoid arthritis. SA Orthopaedic Journal 2010; vol.9, no. 2, pp.
30-33.
CONFERENCE PROCEEDINGS
Peer-reviewed published abstracts:
1. M. M. Ally, B. Hodkinson, P. W. A. Meyer, E. Musenge, M. Tikly, R. Anderson.
Relationships between radiographic damage, shared epitope alleles and
circulating cytokines in Dmard-naïve early RA patients: Annals of the Rheum
Diseases 2013 Jun; 72(Suppl 3): A375-A376 [Annual European Congress of
Rheumatology EULAR abstracts 2013, 12-15 June 2013, Madrid, Spain].
doi:10.1136/annrheumdis-2013-eular.1150.
2. Ally Mahmood MTM, Hodkinson Bridget, Meyer Pieter WA, Musenge
Eustasius, Tikly Mohammed, Anderson Ronald. Matrix metalloproteinase 3
and acute phase proteins as markers of disease activity and radiographic
damage in early rheumatoid arthritis. Arthritis & Rheumatism 2012; 64 Suppl
10:1794. [Abstracts of the American College of Rheumatology/Association of
Rheumatology Health Professionals Annual Scientific Meeting, Washington,
DC, November 9-14, 2012].
5
3. Hodkinson B, Meyer PWA, Musenge E, Ally M, Anderson R, Tikly M. Unique
circulating cytokine profile of rheumatoid arthritis patients with nodules. Journal
of Translational Medicine 2011; 9(Suppl 2): 48. [Meeting abstracts of the 6th
European Workshop on Immune-Mediated Inflammatory Disease, Nice,
France, 23-25 November 2011]
4. Meyer PWA, Hodkinson B, Ally M, Musenge E, Wadee AA, Tikly M, Anderson
R. Circulating anti-CCP antibodies and cytokines as biomarkers of response to
methotrexate in rheumatoid arthritis patients. Immunology 2011; 135(Suppl 1):
233. [Abstracts of the Annual Congress of the British Society for Immunology,
5-8 December 2011, Liverpool, UK].
5. Meyer P, Hodkinson B, Ally M, Musenge E, Wadee AA, Tikly M, Anderson R.
Circulating cytokine profiles and their relationships with rheumatoid factor,
antibodies to citrullinated peptide/vimentin, acute phase reactants, and HLADRB1 shared epitope alleles in patients with rheumatoid arthritis. Annals of the
Rheumatic Diseases 2010; 69(Suppl 3): 180. [Rome, 2010].
6. Hodkinson B, Meyer P, Musenge E, Ally M, Anderson R, Tikly M. Gammainterferon is the dominant cytokine in rheumatoid nodulosis. Annals of the
Rheumatic Diseases 2011; 70(Suppl 3): 689. [London, 2011].
Presentations at biomedical congresses:
1. Ally M, Hodkinson B, Meyer P, Musenge E, Anderson R, Tikly M. Matrix
metalloproteinase-3, a marker of disease activity and radiographic change in
early RA. Congress of the South African Rheumatism and Arthritis Association,
9-11 September 2011, Johannesburg. (Oral presentation).
2. Ally Mahmood MTM, Hodkinson Bridget, Meyer Pieter WA, Musenge
Eustasius, Tikly Mohammed, Anderson Ronald. Matrix metalloproteinase 3
and acute phase proteins as markers of disease activity and radiographic
damage in early rheumatoid arthritis. Arthritis & Rheumatism 2012; 64 Suppl
10:1794 [Abstracts of the American College of Rheumatology/Association of
Rheumatology Health Professionals Annual Scientific Meeting, Washington,
DC, November 9-14, 2012]. (Poster presentation).
3. M. M. Ally, B. Hodkinson, P. W. A. Meyer, E. Musenge, M. Tikly, R. Anderson.
Relationships between radiographic damage, shared epitope alleles and
circulating cytokines in Dmard-naïve early RA patients: Ann Rheum Dis
2013;72:Suppl 3 A375-A376. EULAR 12-15 June 2013, Madrid, Spain.
doi:10.1136/annrheumdis-2013-eular.1150. (Poster presentation).
6
4. Hodkinson B, Meyer P, Musenge E, Ally M, Anderson R, Tikly M. Gammainterferon is the dominant circulating cytokine in rheumatoid nodulosis.
Congress of the European League Against Rheumatism, 6-9 June 2011,
London, UK. (Poster presentation).
5. Hodkinson B, Steel HC, Meyer P, Musenge E, Ally M, Anderson R, Tikly M.
Unique circulating cytokine profile of rheumatoid arthritis patients with nodules.
Sixth European Workshop on Immune-mediated Inflammatory Diseases, 23-25
November 2011, Nice, France. (Poster presentation).
6. Meyer PWA, Hodkinson B, Ally M, Musenge E, Wadee AA, Tikly M, Anderson
R. Circulating anti-CCP antibodies and cytokines as biomarkers of response to
methotrexate in rheumatoid arthritis patients. Annual Congress of the British
Society for Immunology, 5-8 December 2011, Liverpool, UK. (Poster
presentation).
7. Hodkinson B, Meyer P, Musenge E, Ally M, Anderson R. Gamma-interferon is
the dominant circulating cytokine in rheumatoid nodulosis. Congress of the
South African Rheumatism and Arthritis Association, 9-11 September 2011,
Johannesburg. (Oral presentation).
8. Hodkinson B, Meyer P, Musenge E, Ally M, Anderson R, Tikly M. Functional
disability and health-related quality of life in indigent South Africans with early
rheumatoid arthritis. Congress of the South African Rheumatism and Arthritis
Association, 9-11 September 2011, Johannesburg. (Oral presentation).
9. Meyer P, Hodkinson B, Ally M, Musenge E, Wadee AA, Fickl H, Tikly M,
Anderson R. New shared epitope classification and its relationship to RAassociated autoantibodies, circulating biomarkers and clinical indices.
Congress of the South African Rheumatism and Arthritis Association, 9-11
September 2011, Johannesburg. (Oral presentation).
10. Hodkinson B, Ally M, Musenge E, Meyer P, Anderson R, Tikly M. Low level of
formal education yields poorer outcomes in early rheumatoid arthritis patients
in South Africa. Congress of the African League Against Rheumatism, 5-7 May
2011, Algiers, Algeria. (Oral presentation).
11. Hodkinson B, Ally M, Musenge E, Meyer P, Anderson R, Tikly M. Response to
traditional disease-modifying anti-rheumatic drugs in indigent South Africans
with early rheumatoid arthritis.Congress of the African League Against
Rheumatism, 5-7 May 2011, Algiers, Algeria. (Oral presentation).
7
TABLE OF CONTENTS
Page
Declaration
2
Acknowledgements
3
Publications
4
Table of Contents
8
Abbreviations
15
Synopsis
19
CHAPTER 1
1.1
Introduction
22
1.1.1
The Immune Response
22
1.1.2
Cytokines and intracellular signalling domains
24
1.1.3
Cytokine receptors
25
1.1.4
Key cytokines and their signalling pathways
27
1.1.4.1 Pro-inflammatory
27
1.1.4.2 Anti-Inflammatory
30
1.2
Pathogenesis of rheumatoid arthritis
36
Overview
36
1.2.1
Genetics of rheumatoid arthritis
38
1.2.2
Non-HLA genes
39
1.2.3
Epigenetic factors
40
1.2.4
Non genomic factors
41
1.2.5
Synovial Inflammation in RA
43
1.2.6
Immune Dysregulation
44
1.2.7
Cellular Proliferation
46
1.2.8
Disease perpetuation
48
1.2.9
Tissue damage
51
1.3
Biomarkers in RA
53
8
1.3.1
Clinical measures and acute phase response measures
53
1.3.2
Imaging markers
54
1.3.3
Genotyping
55
1.3.4
Autoantibodies
56
1.3.5
Novel laboratory markers
58
1.3.6
Comment
66
1.4
Rheumatoid arthritis in the developing world
68
References
70
CHAPTER 2
Patients and Methods
95
2.1
Study design
95
2.2
Clinical and radiographic methods
96
2.2.1
Disease activity variables assessed
97
2.2.2
Functional assessment included
97
2.2.3
Radiology
97
2.3
Serum
97
2.3.1
Rheumatoid Factor (RF)
98
2.3.2
Anti-citrullinated peptide antibodies (ACPA)
98
2.3.3
Antibodies to modified citrullinated vimentin (anti-MCV)
98
2.3.4
Genotyping
99
2.3.5
Cytokines
100
2.3.6
Serum matrix metalloproteinase-3 (MMP-3) and cartilage
oligomeric matrix protein (COMP)
101
2.4
Treatment
101
2.5
Statistical Analysis
102
2.6
Ethics
102
2.7
Funding obtained from
103
References
104
9
CHAPTER 3
Serum matrix metalloproteinase-3 (MMP-3) in comparison with
acute phase proteins as a marker of disease activity and
radiographic damage in early rheumatoid arthritis
3.1
Introduction
105
3.2
Patients and Methods
107
3.2.1
Patients
107
3.2.2
Laboratory Methods
108
3.2.2.1 Autoantibodies, acute phase reactants, cytokines/chemokines
108
3.2.2.2 MMP-3
108
3.2.2.3 Cartilage oligmeric matrix protein (COMP)
109
3.2.2.4 Share epitope (SE) genotyping
109
3.2.2.5 Statistical Methods
109
3.3
110
3.3.1
3.3.2
3.3.3
3.3.4
Results
Demographic, clinical and laboratory data (autoantibodies, acute
phase reactants, haemoglobin, ESR, MMP-3 and COMP)
Correlations of MMP-3 with clinical indices of disease activity
and non-cytokine, inflammatory biomarkers
Correlations of MMP-3 with cytokines/chemokines/growth
factors
Correlations of COMP with clinical indices of disease activity and
non-cytokine inflammatory biomarkers
110
110
110
111
3.3.5
Association of MMP-3, SAA, CRP and COMP with SE
111
3.3.6
Correlations of CRP and SAA with clinical and laboratory indices
of disease activity
111
3.4
Discussion
111
References
118
CHAPTER 4
Circulating anti-citrullinated peptide antibodies, cytokines, matrix
metalloproteinase-3 (MMP3-) and cartilage metabolites as
biomarkers of response to disease-modifying anti-rheumatic
drug therapy in early rheumatoid arthritis
4.1
Introduction
126
10
4.2
Patients and methods
128
4.3
Statistical analysis
129
4.4
Results
130
4.5
Discussion
132
4.6
Conclusions
136
References
143
Conclusion
155
Appendix 1
158
Appendix 2
165
11
LIST OF TABLES
Page
CHAPTER 1
1
ACR/EULAR 2010 classification criteria for RA
67
CHAPTER 2
1
1987 American College of Rheumatology (ACR) classification
criteria for rheumatoid arthritis
2
Shared Epitope classification according to amino acid sequences
at positions 70-742
96
100
CHAPTER 3
1
Demographic, clinical and laboratory data for the group of RA
patients (n=128)
115
2
Correlations of MMP-3 with indices of disease activity and noncytokine biomarkers
116
3
Correlations of MMP-3 with cytokines
116
4
Correlations of SAA and CRP with clinical and laboratory indices
of disease activity
117
CHAPTER 4
1
SDAI categories
136
2
SDAI measures of response
136
3
Demographics of patients (n=140)
137
4
Significant changes in SDAI scores and circulating biomarker
concentrations following 6 months DMARD therapy
137
5
Subgroup analysis of changes in SDAI values and circulating
biomarkers following 6 months DMARD, for the methotrexate
(MTX) monotherapy and methotrexate-prednisone combination
therapy groups
138
12
6
Significant changes in circulating biomarker concentrations,
stratified according to SDAI at 6 months
138
7
Changes in SDAI scores and circulating biomarkers, stratified by
shared epitope (risk allele) status
140
8
Changes in SDAI and circulating biomarker concentrations
following 6 months of sDMARD therapy
141
13
LIST OF FIGURES
Page
CHAPTER 1
1
Origins and functions of major cytokines
25
2
Activation of NFκB by TNF
27
3
Complement pathways
34
4
Cytokine networks and cellular interactions in cartilage
destruction in rheumatoid arthritis
38
5
RA synovial fibroblast
41
6
Cytokine networks in RA
44
7
Cellular targets of TNF
48
8
Roles of fibroblasts in RA
50
9
Citrullination and carbamylation
58
10
Pathogenesis of RA
59
11
T lymphocyte subset activation within atherosclerotic plaque
64
12
Venn diagram
65
14
ABBREVIATIONS
A
Ab
ACPA
ACR
Ag
AP
APC
ASC(PYCARD)
B
C
D
E
F
ATP
Anti-CarP
BAFF
BLyS
BMD
CCLCCP
CCRCD
COMP
COX-2
CQ
CRP
CTLACTX
CV
CXCLDAMP
DAS
DC
DMARD
DNA
EBV
ECM
EDTA
ELISA
EGF
ESR
FDC
Antibody
Anti-cyclic citrullinated peptides antibodies
American college of rheumatology
Antibody
Activator protein
Antigen presenting cell
Apoptosis-associated speck-like protein containing a
CARD
Adenosine tri-phosphate
Anti-carbamylated protein
B cell activating factor
B Lymphocyte stimulator
Bone mineral density
Chemokine (C-C motif) ligand Cyclic citrullinated peptides
Chemokine receptor Cluster of differentiation
Cartilage oligomeric matrix protein
Cyclo-oxygenase 2
Chloroquine
C-Reactive protein
Cytotoxic T-lymphocyte Antigen Carboxy-terminal collagen crosslinks
Cardiovascular
C-X-C motif chemokine Damage associated molecular pattern
Disease Activity Score
Dendritic cell
Disease modifying anti-rheumatic drug
Deoxy nucleic acid
Epstein barr Virus
Extracellular matrix
Ethylene diamine tetra acetic acid
Enzyme-linked immunosorbent assay
Epidermal growth factor
Erythrocyte sedimentation rate
Follicular dendritic cells
15
G
H
I
J
L
M
FEIA
FGF
FLS
GCs
G-CSF
GM-CSF
GP
GREAT
HDA
HIV
HLA
HSP
IC
ICAM-1
IFN
IgIHD
ILIMT
IP
JAK
LDA
LPS
LT
LV
MAPK
MBDA
MCP
MCP-1
M-CSF
MCV
MDA
MHC
MIP-1
MiRNA
MMP
mRNA
MRI
MTP
Fluorescence immune assay
Fibroblast growth factor
Fibroblast-like synoviocytes
Germinal centres
Granulocyte colony-stimulating factor
Granulocyte and macrophage colony-stimulating factor
Glycoprotein
Gauteng Rheumatoid Evaluation Assessment Trial
High disease activity
Human immunodeficiency virus
Human Leukocyte Antigen
Heat shock protein
Immune complexes
Intercellular adhesion molecule 1
Interferon
Immunoglobulin Ischeamic heart disease
InterleukinIntima medial thickness
Interphalangeal
Janus family tyrosine kinases
Low disease activity
Lipopolysaccharides
Lymphotoxin
Left ventricular
Mitogen-activated protein kinase
Multi-biomarker disease activity
Metacarpophalangeal
Monocyte chemoattractant protein 1 (renamed CCL2)
Macrophage colony stimulating factor
Modified citrullinated vimentin
Moderate disease activity
Major Histocompatibility Complex
Macrophage inflammatory protein 1 (renamed CCL4)
Micro ribonucleic acid
Matrix metalloproteinases
Messenger ribonucleic acid
Magnetic resonance imaging
Metatarsophalangeal
16
N
O
P
R
MTX
Mψ
MYD 88
NADPH
NET
NFAT
NF-κB
NK
NLR
NO
NOD
NSTEACS
OPG
PADPAF
PAMP
PBMC
PCR
PDGF
PGPIP
PTPN22
RA
RANK
RANKL
RANTES
S
RASF
RF
RNA
RNS
ROS
rSSO
SAA
SAPE
SDAI
SE
SENS
SLE
Methotrexate
Macrophages
Myeloid differentiation primary response protein
Nicotinamide adenine dinucleotide phosphate
Neutrophil extracellular trap
Nuclear factor of activated T-cells
Nuclear Factor kappa B
Natural killer
Nod like receptor
Nitric oxide
Nucleotide-binding oligomerisation domain
Non ST segment elevation acute coronary syndrome
Osteoprotegerin
Peptidylarginine deiminase Platelet-activating factor
Pathogen associated molecular pattern
Peripheral blood mononuclear cells
Polymerase chain reaction
Platelet-derived growth factor
Prostaglandin Proximalinterphalangeal
Protein tyrosine phosphatase non-receptor type 22
Rheumatoid arthritis
Receptor activator for nuclear factor κ B
Receptor activator for nuclear factor κ B ligand
Regulated upon activation, normal T cell expressed, and
secreted
Rheumatoid arthritis synovial fibroblast
Rheumatoid factor
Ribonucleic acid
Reactive nitrogen species
Reactive oxygen species
Reverse sequence specific oligonucleotide
Serum amyloid A
R-phycoerythrin-conjugated streptavidin
Simplified disease activity index
Shared epitope
Simplified erosion and narrowing score
Systemic Lupus Erythematosus
17
T
V
Y
SMAD
SNP
sRANKL
ST
STAT
sTNF-R
SWEFOT
SYK
SZP
TB
TCR
TGFTh0
ThTLR
TLT
TNF
TNFR
TRAF
Treg
VAS
VCAMVEGF
YKL-
Small 'mothers against' decapentaplegic
Single-nucleotide polymorphism
Soluble receptor activator for nuclear factor κ B ligand
Synovial tissue
Signal transducer and activator of transcription
Soluble tumour necrosis factor receptor
Swedish Pharmacotherapy Tiral
Spleen tyrosine kinase
Salazopyrin
Tuberculosis
T cell receptor
Transforming growth factor Naive T helper cells
T Helper cell, type Toll-like receptors
Tertiary lymphoid tissue
Tumour necrosis factor
Tumour necrosis factor receptor
TNF receptor-associated factor
T regulatory cells
Visual assessment score
Vascular cellular adhesion molecule Vascular endothelial growth factor
Human cartilage glycoprotein 39
18
SYNOPSIS
Rheumatoid arthritis (RA) is a chronic inflammatory disorder resulting in joint pain,
stiffness and joint destruction. This can lead to considerable loss of physical and
psycho-social function and even premature death. However, numerous advances in
the management of RA have led to remarkable life-changing effects, especially when
therapy is initiated early. Joint inflammation occurs following an abnormal immune
response to some yet unidentified agent. The inciting agent or process is not well
understood but exceptional advances have been made in characterising the
molecular and cellular components of the ensuing inflammatory response. This has
culminated in the identification of novel therapies and potential biomarkers to
enhance patient care. Genetic and environmental factors play an important role with
varying clinical manifestations and unpredictable outcomes on an individual basis.
Globally, RA accounts for considerable morbidity and of particular concern in the
developing world, is the lack of resources that may adversely affect outcome. There
is a paucity of research on various aspects of RA in the developing world.
This study will review immune-pathogenic aspects of RA, the potential use of clinical
features and serological biomarkers and report on aspects of the analysis of a
prospective observational study of patients with early RA, prior to starting and 6
months following therapy.
A cohort of 171 early RA patients (disease duration less than 2 years) was recruited
from the rheumatology clinics of two tertiary academic hospitals, located in Gauteng,
South Africa. The objective of the study was to review clinical, demographic,
radiological and serological aspects over a two year period. Outcome measures of
this study included; i) characterisation of disease burden with respect to both physical
and psycho-social aspects; ii) response of disease to therapy in routine care; iii) to
characterize auto-antibody, cytokine, cartilage metabolites and genotype profiles
associated with disease activity and response to therapy; and iv) clinical and
serological measures associated with radiographic damage.
19
Clinical measures included the simplified disease activity index (SDAI) and functional
scores (HAQ-DI and SF-36). At baseline and 6 months rheumatoid factor (RF), Anticitrullinated peptide antibodies (ACPA), circulating cytokines and growth factors were
measured. Cartilage metabolites COMP and matrix metalloproteinase (MMP)-3 were
measured only at baseline. Genotyping for the shared epitope (SE) HLA-DRB1 allele
was performed and classified according to the du Montcel classification. Radiographs
of the hands and feet were taken at baseline and at yearly intervals and evaluated for
erosive disease and scored according to the Larsen grading system.
The cohort recruited was mostly female (140) with a mean age of 47 years. Despite a
mean symptom duration of 12 months, patients had severe disease with a mean
SDAI of 39.4, with nodulosis and erosive diseases seen in 23% and 51 %
respectively. Auto-antibody tests revealed high sensitivity (83%) and specificity (85%)
for ACPA with the highest specificity (95%) if both RF and ACPA were positive.
Shared epitope positivity was found in 92% of patients and correlated strongly with
ACPA. Cytokine levels were globally increased favouring a T helper cell (Th)1,
macrophage and pro-angiogenic cytokine profile in certain sub-groups. MMP-3 levels
correlated with measures of disease activity but did not perform better than routine
laboratory measures such as the C-reactive protein (CRP).
Serial measurement of auto-antibodies and cytokine profiles at 6 months revealed
significant decreases in tandem with a decrease in disease activity. A more robust
decline in cytokine levels was noted in patients who achieved a low disease activity
state (SDAI<11) and in patients who received prednisone in addition to methotrexate
(MTX). Of the auto-antibodies and biomarkers tested only MMP-3 was predictive (OR
2.8) of an SDAI>11 at 6 months.
Only 28% of patients achieved a low disease activity state at 12 months. Predictors of
outcome at 12 months included low level of education, unemployment, radiographic
damage and a high disease activity.
20
Conclusion: RA in sub-Saharan Africa is strongly associated with SE and ACPA
status and a high disease burden at presentation. Despite therapy, only a small
percentage of patients achieved an acceptable level of disease activity in routine care
underscoring the need for a more aggressive therapeutic approach. Serial
measurement of auto-antibodies and cytokines had limited clinical value in routine
patient care, but may guide future therapies with the potential to help in stratifying
patients. The measurement of serum MMP-3 at baseline may identify a subgroup of
patients with a poorer prognosis and hence allow for a more intensive therapeutic
approach from the outset.
21
CHAPTER 1
1.1
Introduction
Rheumatoid arthritis is a chronic inflammatory arthritis of unknown aetiology. An
autoimmune pathogenic mechanism is central to disease initiation and perpetuation,
with inflammation and tissue destruction resulting in a vicious cycle, leading to
disease persistence and progression.1 Recent insights into the molecular pathways
characterizing these pathogenic processes have not only led to numerous innovative
therapies, but have also provided a better understanding of the immune-inflammatory
response. RA is an archetypal autoimmune disease, which is associated with other
physiological and pathological conditions such as aging, osteoporosis, cardiovascular
disease and malignancies.
1.1.1 The Immune Response
The innate and adaptive systems are two components of the immune system which
act in concert to defend against foreign infections and promote healing following an
injury. The innate response is the initial non-specific response to foreign antigen
comprising cells of the macrophage monocyte lineage, dendritic cells, neutrophils,
and complement. Macrophages and dendritic cells not only phagocytose / pinocytose
foreign agents but also process the antigen for presentation and activation of the
adaptive immune system. Macrophages demonstrate remarkable plasticity depending
on the nature of the stimulus, resulting in anti- or pro-inflammatory responses.
Dendritic cells share many characteristics of macrophages and are key antigen
presenting cells, establishing an important link with the adaptive immune response.2
Surface and cytosolic toll like receptors (TLR), as well as cytosolic nucleotide-binding
oligomerisation domain (NOD) like receptors (NLR) of dendritic and other antigen
presenting cells, recognize and interact with numerous pathogen-associated
molecular patterns (PAMPs) and damage-associated patterns (DAMPs). The
adaptive response is a delayed, but specific response, comprising of T and B
lymphocytes. The adaptive response is initiated by antigen presentation and
22
activation of naive T cells. Depending on the stimulus and host environment, T cells
differentiate into Th1 (cell-mediated response), Th2 (humoral /antibody-mediated
response), Th17 (pro-inflammatory response) and regulatory T cells (Tregs)
(inhibitory response). T cell activation results in a specific response and retention of
memory to a particular antigen.
A crucial host factor is the major histocompatibility complex (MHC) or human
leukocyte antigen (HLA) system which is encoded by the chromosomal area
important for self-recognition, tolerance and displaying of antigens to T cells. The
MHC is classified into 3 classes I, II and III. Class I comprises three groups A, B, C
and class II is a single group, D, that is further subdivided into three, DP, DQ, and
DR. Class I is found on all nucleated cells and plays a key role in self-tolerance and
protection against viral infections. Class II is displayed on antigen presenting cells
involved in regulating the immune response. Class III consists of genes encoding
complement proteins and certain cytokines.
Functional changes to DNA without alterations in structure occur through biochemical
processes resulting in epigenetic modulation of protein transcription. These
biochemical changes include methylation and acetylation that can influence gene
expression. Epigenetic changes may also occur following coupling and uncoupling of
chromosomal nucleotides with certain polypeptides such as ubiquitin and ubiquitinlike peptide.3
Cytokines are molecular messengers which play an integral role in the cross-talk
between various components of the immune response and host, allowing for a coordinated and regulated response. Numerous intracellular signaling domains respond
to these molecular signals and modulate the cellular response. Genetic factors
including genotype and epigenetic changes often influence the ultimate cellular
response.
Cellular and molecular components of the immune-inflammatory response are subject
to intensive research in the field of rheumatology. An ever-expanding knowledge of
23
these networks has led to an in-depth understanding of pathogenic mechanisms,
potential biomarkers and the development of novel therapies.
A more detailed review of some of these components follows.
1.1.2 Cytokines and intracellular signaling domains
Cytokines are molecular messengers which promote communication between various
components of the immune response, working in an autocrine and paracrine manner.
Functional heterogeneity is the order of the day with tremendous plasticity and
elements of redundancy. Components can be classified according to predominant
functional responses (pro-inflammatory/anti-inflammatory), structural similarities, cells
of origin (Th1/Th2/Th17) or production by innate or adaptive immunity. The origins
and functions of major cytokines are shown in Figure 1.
24
Fig 1. Antigen presentation by DCs to naive T cells and other factors (innate immune response
substances, vitamins, cytokines in the environment) induces the T cells to produce ILs and differentiate
into TH1, TH2, TH9, TH17, TH22, or follicular TH (TFH) cells. These T-cell subsets can promote
different types of inflammatory responses on the basis of their respective cytokine profiles, responses
to chemokines, and interactions with other cells.from : J Allergy Clin Immunol.
2011; 127(3):701-721).[with permission]
Cytokines mediate their effects through receptors followed by intracellular signal
transduction and gene transcription. This affects gene expression, cell survival and
movement.
1.1.3 Cytokine receptors
There are 5 different families of receptors based on differences in their structures: i)
the four alpha helix bundle cytokine family, ii) Tumour necrosis factor (TNF) receptor
family, iii) Transforming growth factor (TGF) family, iv) Interleukin (IL)-1 family and v)
IL-17 family.4
25
Intracellular signaling is mediated by membrane bound or cytosolic domains
consisting of enzymes and structural proteins; once activated they initiate a cascade
of chemical or conformational changes that activate transcriptional factors;
intracellular changes include protein phosphorylation, ubiquitination, sumoylation,
degradation and translocation.5 Phosphorylation is often mediated by kinases while
ubiquitination involves the attachment of a 76 amino acid protein called ubiquitin.
Sumoylation is related to the interaction with a ubiquitin-like peptide. Some of the
intracellular protein complexes include kinases, adaptor proteins, and TNF receptorassociated factor (TRAF). Transcriptional proteins include nuclear factor kappa B
(NFκB), signal transducer and activator of transcription (STAT), nuclear factor of
activated T-cells (NFAT) and activator protein (AP)-1, culminating in the expression of
the relevant effector gene. Intracellular signaling is essential not only for the response
to cytokines, but also for activation or modulation by cell-cell contact, and responses
to co-stimulatory ligands and antigenic peptides (Figure 2). Several different
extracellular signals share the same signal transduction pathways or transcription
proteins, contributing to the complexity and redundancy seen.
26
Fig 2. Activation of NF-κB by TNF - Signal transduction through TNF receptor domains such as
TRAF 2 result in NFκB mediated transcription of genes by TNF. Intracellular processes include
phosphorylation,ubiquitination and sumoylation: from Jürgen Ruland Nature Immunology12,709–
714(2011)[with permission]
Clinical trial outcomes in patients with various autoimmune diseases, using
therapeutic modalities targeting components of the immune-inflammatory response
underscore their involvement as dominant role players in the immune response.
1.1.4 Key cytokines and their signaling pathways
1.1.4.1
Pro-inflammatory
IL-1 beta
IL-1 beta is important in the defense against pathogens, especially intracellular
microorganisms such as Mycobacterium tuberculosis; however, elevated levels are
also found in many pathological conditions. Signal transduction occurs after binding
to surface IL-1 R1 receptors, resulting in synthesis of an inactive pro-form through
27
activation of the NFκB pathway following interaction with the adaptor protein myd88,
several kinases and TRAF 6. The inactive pro-form is activated by the enzyme
caspase 1. Caspase 1 requires activation through a protein complex called the
inflammasome.6 Inflammasome activation is promoted by a decrease in intracellular
potassium levels related to increased levels of adenosine triphosphate (ATP)
released by macrophages extracellularly.7 Stimulation of antigen presenting cells by
PAMPs or DAMPs can also result in the activation of caspase 1 through interactions
with the inflammasome adaptor protein, apoptosis-associated specklike protein
[ASC(PYCARD)].6 IL-1 activation results in a host of inflammatory actions, T and B
cell differentiation, and production of other cytokines such as IL-6 and IL-17.8 The
effects of IL-1 beta are kept in balance by the production of IL-1Ra and the presence
of decoy, biologically-inert IL-1 receptor type II (IL-1R II).
IL-6
This is a key cytokine involved in hematopoiesis, the acute phase response and
inflammation. Signal transduction is mediated through a cell surface complex of IL-6
and its receptor Glycoprotein (GP) 130. This then undergoes phosphorylation by
Janus family tyrosine kinases (JAK), followed by transcription via STAT 3. IL-6
expression is regulated by negative feedback of intracellular signaling pathways and
transcription. IL-6 is produced by many cells with endothelial cells, monocytes/
macrophages and fibroblasts being the key producers during inflammation. IL-6
promotes neutrophil movement, macrophage maturation, B cell antibody isotype
switching and differentiation into plasma cells. IL-6 also promotes inflammation by
inducing Th17 cell differentiation from naïve Th (Th0) cells and Treg cells.9,10
IL-7
IL-7 regulates T cell proliferation and cytokine production [interferon (IFN)-γ and
TNF], driving a Th1/Th17 response, as well as cytotoxic CD 8+ T cell and natural killer
(NK) cell proliferation.
IL-7 also contributes to differentiation of naïve and memory B cells. After binding to its
surface receptor IL-7Rα, intracellular signaling pathways include JAK 3 and STAT
transcription.11
28
IL-8
This cytokine belongs to the sub-family of chemotactic cytokines termed chemokines;
the primary role of IL-8 is chemotaxis of neutrophils, NK cells, and T cells, as well as
in angiogenesis at sites of inflammation. IL-8 transcription is a rapid process following
activation of mitogen-activated protein kinases (MAPK), with NFκB and AP-1 as
transcription factors.9
IL-17
IL-17 and its corresponding receptor IL-17R form a distinct group with a diverse set of
pro-inflammatory manifestations through MAPK/NFκB intracellular signaling. IL-17
comprises of 5 subtypes A to E with IL-17A being the predominant
subtype.Production is predominantly from a specific T cell subset called Th17 cells,
but is also produced from a host of other cells such as CD 8+ T cells, NK cells,
neutrophils, fibroblasts, endothelial cells and B cells. IL-17 effects are mediated by
inducing the release of other cytokines such as IL-1, IL-6, IL-8, TNF, granulocyte and
macrophage colony stimulating factor (GM-CSF), granulocyte colony stimulating
factor (G-CSF), and monocyte chemoattractant protein (MCP)-1 as well as matrix
metalloproteinases (MMPs). IL-17 plays a crucial role in host defense against
extracellular pathogens such as Klebsiella pneumoniae, Toxoplasma gondii,
Porphyromonas gingivalis and Candida albicans.9
IFN-γ
This cytokine is produced by cells of both the innate and adaptive components of the
immune response. IFN-γ mediates its effects via binding to 2 cell surface receptors
and JAK/STAT signal transduction. It is produced by macrophages, NK cells, Th1 and
B cells. Production is stimulated by cytokines such as IL-12 and IL-18 from APC,
while inhibition of synthesis is mediated by IL-4, IL-10, TGF-β and glucocorticoids.
IFN- γ primarily drives a Th1 response and inhibits Th2 differentiation. Augmentation
of the inflammatory response occurs through regulation of endothelial leukocyte
interaction and upregulation of chemokine production and adhesion molecule
expression. Upregulation of MHC class I molecules by IFN-γ facilitates eradication of
29
intracellular pathogens, while upregulation of MHC class II in APC promotes CD4+ T
cell activation.9
TNF
Inhibitors of TNF represent one of the most successful class of biologic agents in use
clinically for immunotherapy of immune-mediated inflammatory diseases. This
underscores the importance of this cytokine in the immune inflammatory response.
There are numerous cell surface and circulating soluble receptors [tumour necrosis
factor receptor (TNFR)] regulating cell activation. Depending on the stimulus and
intracellular signaling pathway, TNF mediates cellular proliferation, secretion of
cytokines and apoptosis.12 The resultant stimulation of both the innate (macrophages,
NK cells) and adaptive immune systems (T and B cells) results in TNF production.
Signal transduction through TNF receptor domains such as TRAF 2 and MAPK result
in NFκB mediated transcription of genes, augmenting inflammation via induction of
cytokines such as IL-1, IL-6, which in turn promote neutrophil chemotaxis,
macrophage activation and adhesion molecule expression.
1.1.4.2
Anti-Inflammatory
IL-10
This cytokine is produced predominantly by Tregs and is a potent inhibitor of
inflammation inhibiting the secretion of numerous cytokines and chemokines. It is
also produced by B cells, macrophages and dendritic cells. IL-10 decreases the
expression of surface MHC class II molecules on APC and inhibits T cell costimulation. Intracellular signaling following Il-10/IL-10R binding is via the JAK/STAT
pathway.9,13
TGF-β
Secreted TGF-β circulates as a large inactive complex that requires proteolytic
activation by plasmin, MMPs and thrombospondin 1 to bind to its surface receptors.
Potent immune-regulatory effects are then mediated by various kinases and small
‘mothers against’ decapentaplegic (SMAD) transcription factors. Inhibitory effects
30
include prevention of NFκB translocation into the nucleus, thus downregulating the
expression of numerous pro-inflammatory cytokines.14 TGF-β promotes B cell isotype
switching to immunoglobulin (Ig) A production. Secreted predominantly by Tregs,
TGF-β inhibits Th1and Th17 cell differentiation. It is also secreted by macrophages
and dendritic cells to, acting negative feedback control.
Dendritic cells
These are cells of the innate immune system with branch-like projections which play
an important role in self-tolerance and the immediate immune response to foreign
antigen. These cells originate from haematopoietic stem cells and circulating
monocytes. Three different types have been characterized: i) classical; ii)
plasmacytoid; and iii) inflammatory type.
The classical dendritic cells are resident in the interstitial space, skin (Langerhans
cells), spleen and lymphoid organs. Dendritic cells are also found in the lungs and
gastro-intestinal tract, but are absent in the brain parenchyma with microglia fulfilling
this function.15 Plasmacytoid dendritic cells play an important role in defense against
viruses though the immediate release of type I interferon (IFN-I). Inflammatory
dendritic cells concentrate at sites of inflammation and release mediators such as
TNF and reactive oxygen species.16 Dendritic cell activation requires interaction with
antigen through non-specific receptors, such as Toll-like receptors (in the membrane
and cytosol) and NOD-like receptors (cytosolic). As mentioned earlier, these
receptors recognise not only PAMPs, but also damaged endogenous components
(DAMPs). Antigen uptake and processing promotes presentation of the antigen to the
adaptive immune system. These cells are also capable of migrating to draining lymph
nodes through the action of certain chemokines, enabling the stimulation of the
adaptive immune response. The degree of response and type of T cell differentiation
depends on antigenic stimulus, dendritic cell subset and cytokine profile. Dendritic
cells mediate some of these effects through up regulation of adhesion molecules, costimulatory ligands and the secretion of a host of cytokines such as TNF, IL-2, IL-4,
IL-6, IL-10, IFN-γ and TGF-β.17
31
Macrophages
Together with dendritic cells, macrophages are key players in antigen processing and
presentation to the adaptive immune system. Macrophages originate from cells of the
monocyte lineage and influence numerous physiological and pathological processes
effected predominantly by phagocytosis and cytokine release. Macrophage
differentiation in the bone marrow is predominantly controlled by two colony
stimulating factors, macrophage colony stimulating factor (M-CSF) and GM-CSF, with
further differentiation influenced by antigenic stimulus and host factors, resulting in
phenotypes expressing different functions of tissue repair, host defense and immuneregulation.11 These different functions can be classified into two subtypes M1 (proinflammatory) and M2 (anti-inflammatory or homeostatic). M1 macrophages are
activated by lipopolysaccharides (LPS), IFN-γ, TNF and GM-CSF; alternatively M2
macrophages are stimulated by IL-4, IL-10, IL-13, immune complexes and M-CSF.
Activated M1 macrophages release reactive oxygen species (ROS), nitric oxide (NO),
IL-6, and TNF, while upregulating surface receptors and adhesion molecules; in
addition they play an important role through phagocytosis, clearing foreign antigens,
debris and apoptic material.18
Neutrophils
These cells are involved in the initial response to bacterial and fungal infections.
Movement of neutrophils to sites of inflammation requires migration through the
endothelium enabled by various chemokines and adhesion molecules. Surface
receptors such as Fc- receptors, β2 integrins and selectins mediate neutrophil
adhesion and responses via intracellular signaling using specific kinases such as
spleen tyrosine kinase (Syk). Notwithstanding generation of ROS numerous enzymes
capable of degradation of proteins and cell wall components are present in neutrophil
granules. Another mechanism which protects against foreign pathogens is a distinct
form of ‘programmed’ cell death resulting in release of net-like fibers, a process called
NETosis. These fibers contain DNA, histones and components of neutrophil granules.
These extracellular fibers trap and kill bacteria. Neutrophil extracellular trap (NET)
formation can be stimulated by bacterial LPS, ROS, IL-8, platelet activating factor
32
(PAF), C5a, GM-CSF, IFN-α and -γ. An important step in NET induction is the
generation of ROS, resulting in citrullination of arginine in histones via
peptidylarginine deiminase (PAD)1. As nuclear material is also released within the
extracellular traps the neutrophil dies.19
Neutrophils also produce cytokines that promote further recruitment of neutrophils
amplifying the inflammatory response.
Complement
The complement system is an integral component of the innate immune system
consisting of circulating and cell-bound proteins and glycoproteins. Components are
produced mainly in the liver, but are also produced by cells of the monoctyte lineage.
Complement proteins act in a domino-like manner, culminating in opsonization and/or
lysis of target pathogens. Three different activating pathways classical, alternative
and lectin-binding, are induced by different antigens in the presence or absence of
antibodies. Antigenic stimulants of the three pathways include:
i)
Classical antigen/antibody (Ag/Ab) complexes, nucleic acids, chromatin,
cytoplasmic and mitochondrial components, cardiolipin, CRP, amyloid,
bacterial cell wall components and synthetic particles such as polymers.
ii)
Alternate-carbohydrate moieties on bacteria, fungi, parasites and Ag/Ab
complexes.
iii)
Lectin- carbohydrate moieties (mannan-binding lectin) found on many
microorganisms.
All three pathways result in activation of C3 and C5, culminating in the formation of
the membrane attack complex (MAC), forming a pore in the target cell membrane
initiating cell lysis20 (Figure 3). Activated C3 and C5 are potent mediators of
inflammation and opsonization; additionally C5 promotes chemotaxis of neutrophils
via generation of C5a.16 These events are summarized in Figure 3.
33
Fig 3. Complement pathways Diagram outlining the three complement pathways. MBL = mannosebinding lectin; MASP = MBL-associated serine protease; TCC = terminal complement complex; MAC =
membrane attack complex: from http://openi.nlm.nih.gov/ Busche MN, Stahl GL - Ger Med Sci 2010
[open access ]
T cells
The primary event in the stimulation of the adaptive immune response is T cell
activation by the innate immune system. The antigenic stimulus and host factors
determine the pathway of T cell differentiation, viz. CD4+ T helper 1, 2 or 17, or CD 8+
T cytotoxic lymphocytes. Antigen presentation in an IL-12 cytokine milieu promotes
Th1 development, resulting in cell-mediated immunity against intracellular
microorganisms through release of cytokines such as IFN-γ and IL-2. These events
are counter balanced by production of the inhibitory cytokine IL-10.
Antigen presentation in an IL-4 milieu facilitates Th2 differentiation with production of
IL-4, IL-5, IL-10, and IL-13, allowing for antibody or humoral immunity. This pathway
is particularly important for protection against helminthes and certain protozoa.
Activation of T cells also requires co-stimulation with antigen presenting cells (APCs)
34
through interaction of CD28 on T cells and CD80/CD86 on APC. Th1 inhibits Th2
differentiation via IFN-γ and vice versa Th2 inhibits Th1 via the production of IL-4.
Th17 responses are initiated by an IL-1, IL-6, IL-21, IL-23, TNF and TGF-β milieu.
The Th17 response plays a role in the defense against extracellular Gram-negative
bacteria and Candida infections.
TGF-β plays an important role in keeping the inflammatory response in check by
promoting differentiation of Treg cells.
Dysregulated T cell differentiation results in a host of autoimmune or chronic
inflammatory conditions such as RA (Th1/Th17) and systemic lupus erythematosus
(SLE) (Th2).21
B cells
The primary function of these cells in the immune response is to produce antibodies
which promote clearance of antigens. B cells also have a potent immune-modulatory
function in both the innate and adaptive components of the immune system. Activated
T cells bind to antigen-presenting B cell receptors promoting B cell differentiation to
produce cytokines and specific antibodies.
B cells originate from marrow B lymphocyte precursors that differentiate into different
stages of maturity and can be identified by surface markers called cyclo-oxygenase
(COX)-2 of differentiation (CD) markers. Transitional and mature B cells are APCs
and produce cytokines such as IL-1, IL-6, TNF and counter-regulatory IL-10 and
TGF-β. B cell survival in the peripheral circulation requires the specific cytokine
BAFF/ BLyS (B cell activating factor /B lymphocyte stimulator).
Some B cells are capable of generating antibodies independently of T cell help.
These are innate-like B cells which are able to produce antibodies via interactions
with antigens early in the immune response, producing so-called natural antibodies
protective against pathogens. These innate B cells are also self-reactive, and are
35
thus potentially implicated in autoimmune disease pathogenesis .22 IL-10 production
by B cells is an important regulatory cytokine mechanism, decreasing secretion of
TNF, and IL-1, as well as APC co-stimulatory molecule expression.23
Mature B cells differentiate into professional Ab-producing plasma cells or into B
memory cells, the latter responding quickly on re-challenge with the same antigen.
Five isotypes of antibodies are produced by B cells viz. IgA, IgD, IgE, IgG and IgM,
with isotype shifting dependent on both the antigenic stimulus and cytokine milieu,
(IL-4, IL-13→ IgG and IgE/ TGF-β→ IgA/ IL-10 →memory B cell differentiation/ IL21→ IgG).24
Despite the complexities and perceived redundancies within the immune response a
coordinated and efficient response occurs, resulting in maintenance of normal
homeostasis in an always challenging environment. However, when dysregulated
these events may result in a variety of diseases including chronic infections,
autoimmune disorders and malignancies.
1.2
Pathogenesis of Rheumatoid Arthritis
Overview
Immune dysregulation as a consequence of genetic and environmental factors is the
key driver of chronic inflammation in RA. The HLA system is important for immune
tolerance with specific, genetically determined amino acid sequences associated with
certain diseases.25 In RA this sequence is termed the “shared epitope” and is found in
around 80% of patients. Presence of the shared epitope (SE) varies in different ethnic
groups, with some studies showing a lower prevalence in Black African patients;
however, a recent South African study found as many as 83% positive for the SE in
an early RA cohort, similar to that seen in Caucasians.26
The presence of autoantibodies such as rheumatoid factor (RF) and anti-citrullinated
peptide antibodies is evident even before onset of clinical disease.27 However, the
inciting antigen is unknown, with infective agents such as parvovirus B19 and an
36
organism causing gingivitis (Porphyromonas gingivalis) having been implicated as
possible triggers.27 Smoking has recently received much interest in the possible
initiation of a chemical change in lung tissue – a process called “citrullination”.28
Citrullination involves a chemical change in certain proteins containing the amino acid
arginine which is converted to citrulline. This immunogenic citrullinated protein is now
exposed to the immune system and in genetically predisposed individuals’ results in
the generation of anti-citrullinated peptide antibodies (ACPA). Citrullination also
occurs in the rheumatoid synovium where these antibodies, may not only initiate
disease, but might also account for disease persistence.
Inflammation and tissue destruction may contribute to increasing the antigenic load,
resulting in auto-inflammation and ongoing disease. The synovial pannus is
characterized by new vessel formation, activation of cellular and molecular
components of the immune response and abrasive changes to adjacent cartilage and
bone (Figure 4). Joint destruction is mediated predominantly by pro-inflammatory
cytokines such as tumour necrosis factor (TNF), IL-1, IL-6 and IL-17, which stimulate
synovial fibroblasts, chondrocytes and oseoclasts. Bony changes in RA include focal
erosions, peri-articular osteopenia and generalized osteoporosis. Cytokines such as
receptor activator of nuclear factor κB ligand (RANKL) and osteoprotegerin (OPG)
play a key role in bone resorption with elevated MMP activity mediating enzymatic
degradation of cartilage.
Once the immune system is activated disease persistence is maintained by as yet
unexplained mechanisms. Phenotypic changes in synovial cells, particularly synovial
fibroblasts, may play a critical role.29 The synovial pannus acts in an autonomous
almost tumour-like fashion, generating pro-inflammatory cytokines, which result in
synovial proliferation and consequent local and systemic effects.6
37
Fig 4. Cytokine networks and cellular interactions in cartilage destruction in rheumatoid arthritis. This
scheme represents the progressive destruction of the cartilage associated with the invading synovial
pannus in rheumatoid arthritis. As a result of immune cell interactions involving T and B lymphocytes,
monocyte/macrophages, and dendritic cells, several different cytokines are produced in the inflamed
synovium as a result of the influx of inflammatory cells from the circulation and synovial cell
hyperplasia. The upregulation of proinflammatory cytokines produced primarily in the synovium, but
also by chondrocytes, results in the upregulation of cartilage-degrading enzymes, of the matrix
metalloproteinase (MMP) and ADAM with thrombospondin-1 domains (ADAMTS) families, at the
cartilage–pannus junction. Chemokines, nitric oxide (NO), and prostaglandins (PGs) also contribute to
the inflammation and tissue catabolism. SDF, stromal cell-derived factor 1; TNF, tumor necrosis factor;
TGF, transforming growth factor; IFN, interferon; Treg, regulatory T lymphocytes; Th, T helper cells:
from Otero and Goldring Arthritis Research & Therapy 2007 9:220 doi: 10.1186/ar2292)[open access]
1.2.1 Genetics of Rheumatoid Arthritis
Certain genetic studies have reported a concordance of around 15% in monozygotic
twins suggesting other factors may be more significant, but taking into account the
prevalence of RA overall heritability accounts for 40-65% of RA susceptibility.31
Multiple gene loci and epigenetic factors superimposed on environmental factors
influence not only susceptibility, but also severity and response to therapy.
The HLA (human leukocyte antigen) family of genes is one of the most important
genetic determinants of RA. These genes code for proteins expressed almost
38
exclusively on the surface of APC such as macrophages and dendritic cells allowing
for a matching fit with inciting antigen and presentation to CD4+ T cells.16
A common or shared amino acid sequence, RAA (R-arginine, A-alanine), at position
72 to 75 of certain HLA-DR 1 (HLA-DRB1*01) and –DR 4 (HLA-DRB1*04) alleles has
been shown to confer an increased risk for RA.This region, as mentioned above, is
temed the shared epitope (SE) and confers a measure of susceptibility influenced by
two adjacent amino acids at positions 71 and 72. The QKRAA, QQRAA and KKRAA
(Q-glutamine, k-lysine) sequences increase the risk of RA, while the DERAA (Daspartic acid, E- glutamic acid) sequence has a protective effect.32,33 A new
classification based on the amino acid sequences adjacent to the SE has been
validated. Depending on the amino acid sequence, groups are classified as S1, S2,
S3P, S3D and X. The S2 and S3P alleles are associated with increased RA
susceptibility. The low risk alleles S1, S3D and X are pooled together as L alleles.
Certain non SE HLA-DRB alleles (HLA-DRB1*15, HLA-DRB*1301, -DRB*1302,DRB1*1304) have also been shown to be associated with an increased susceptibility
to RA and have been incorporated into the S1 group.34,35
The SE HLA alleles account for 11% to 37% of RA heritability particularly in patients
homozygous for the risk allele.32 The presence of SE increases the risk of extraarticular manifestations and disease progression.
1.2.2 Non–HLA genes
Genome-wide association studies have also implicated a host of other, albeit less
frequent, associations with RA including polymorphisms of molecules involved in:
i)
Intracellular signalling [protein tyrosine phosphatase non-receptor
(PTPN22), TRAF1, STAT4]
ii)
T cell co- stimulation or B cell interaction [CD28, CD40, cytotoxic Tlymphocyte antigen (CTLA4)],
iii)
Cytokines that regulate activation of T cells [IL-2, IL-21, IL-23R, TNF,
Chemokine receptor (CCR) 6]
iv)
Enzymatic conversion of arginine to citrulline (PAD14).
39
Notwithstanding HLA-DRB1, the genes coding for PTPN22 and IL-23R show the
strongest association with RA susceptibility.
1.2.3 Epigenetic factors
Not only the DNA sequence, but also chemical, or so called epigenetic modifications
to genes, modulate the immune response. These changes are heritable and some
environmentally induced changes may be reversible. Chemical changes include DNA
methylation and post translational modification of chromatin structure by acetylation,
phosphorylation, methylation, ubiquitination and sumoylation.36,37
DNA methylation is an important inhibitory signal for gene expression and
hypomethylation has been demonstrated in synovial fibroblasts, certain cytokine
promoter genes (IL-6, IL-10) and micro ribonucleic acid (miRNAs) in patients with RA
(Figure 5). MiRNAs do not code for any peptides, but rather affect gene expression
by blocking the binding site of the target messenger ribonucleic acid (RNA). Abnormal
miRNAs and consequent downstream effects have been demonstrated in RA. In the
rheumatoid arthritis synovial fibroblast (RASF), this results in over-expression of MMP
1, IL-6 and a change to a tumour like phenotype.36,37 In addition to effects on the
RASF, acetylation/ de-acetylation of histones also affects transcription factors such
as NF-kB and secretion of cytokines such as TNF and IL-6.36
40
Fig 5. RA synovial fibroblast: from Nature Reviews Rheumatology 5, 266-272 : May 2009 [with
permission]
1.2.4 Non genomic factors
As mentioned above, smoking is one of the strongest environmental factors
increasing the risk and severity of RA. The risk of RA in smokers increases with
longer duration of smoking and amount smoked. The equivalent of more than a 40
pack year history is associated with doubling of the risk for RA in comparison to nonsmokers. The increased risk remains significant for greater than 20 years after
stopping cigarette smoking.38 Smoking has been associated with a specific sub-type
of RA namely ACPA-positive RA, particularly in SE positive patients. However, there
may be ethnic differences as a study in African Americans did not show an
association between smoking status and ACPA.39 In patients homozygous for the SE,
the risk for RA is increased almost 21-fold increase compared to SE-negative nonsmokers. The initial event may be distal to the joints, possibly in the lungs wherein
smoking has been shown to increase citrullination.38 Citrullinated peptides fit more
easily into the antigen binding sites of APCs, initiating the cascade of events to follow.
41
A whole host of RA-associated antibodies has been described, probably accounting
for the varied clinical phenotype evident in practice. Two well-characterised
autoantibodies, RF and ACPA, are found long before the onset of clinical disease as
mentioned earlier.
RF is an antibody which binds to the Fc portion of IgG. Sero-positivity for RF is
associated with disease severity. The pathogenic role of RF seems to be through
immune complex-mediated complement activation and generation of chemotactic
factors. RF is found in 80 to 90 % of patients with established disease. Most are of
the IgM and IgG isotypes with IgE RF being associated with extra-articular
manifestations and mast cell activation. Routine laboratory assays usually detect IgM
RF.
ACPA are found in about 12-15% of patients who are sero-negative for RF. The
specificity of ACPA for RA is around 90% and even higher if RF is positive. ACPA are
also found long before the onset of disease and a “second hit” such as an infection or
trauma resulting in citrullination of joint tissue may lead to expansion of the affinity of
the initial ACPA autoantibodies, so-called “epitope spreading,” reaching a critical
threshold, culminating in clinical disease. The increase in isotypes occurs just prior to
disease onset. ACPA-positive patients with the highest titers manifest radiographic
progression and RA-associated morbidity. Patients positive for the SE are more likely
to be ACPA positive, with ACPA-negative disease having a different pathogenic
mechanism and phenotype.
Perodontitis associated infection due to Porphyromonas gingivalis results in
citrullination of proteins through a bacterial PAD enzyme. Enolase, a metalloenzyme
involved in glycolysis, when citrullinated, may interact through molecular mimicry with
ACPAs produced in response to citrullinated bacterial enolase.40 Other infections
such as those caused by mycoplasmas, parvovirus B19, retroviruses, mycobacteria,
Epstein Barr virus (EBV) and bacterial cell wall components may initiate the immunopathology seen in RA through stimulation of TLRs. Animal models of arthritis
implicate TLRs 2, 4 and 9 . Mycoplasmas can induce cytokine release from
macrophages initiating or exacerbating arthritis in murine arthritis models.41 EBV may
42
induce arthritis through the mechanism of molecular mimicry as the EBV glycoprot
GP 110 contains the amino acid sequence of the SE, QKRAA. Parvovirus B19 genes
have also been demonstrated in RA synovial tissue, possibly activating RASF.42
Hormonal factors may also account for the female preponderance of RA, as estrogen
has been shown to render autoreactive B cells more resistant to apoptosis while
stimulation of estrogen receptors on fibroblast-like synoviocytes (FLS) and
macrophages increases production of MMPs and TNF. Pregnancy is associated with
remission in around 75% of patients, possibly due to increased expression ofIL-10;
however, 90% of patients experience flares in the post-partum period. The flare is
usually associated with a rise in RF titre. Breast feeding for longer than 13 months
has also been shown to be associated with a decreased risk of developing RA.
The role of red meat, vitamin D, and oral contraceptives in the pathogenesis of RA is
controversial. Studies have demonstrated an inverse relationship between
socioeconomic status and education level with susceptibility of developing RA.38
The interaction of environmental and genetic aspects seems pivotal in the
pathogenesis of RA resulting in chronic inflammation, primarily in the synovium with
ensuing articular and extra-articular manifestations.
1.2.5 Synovial Inflammation in RA
The pathological immune response triggers a cascade of cellular and molecular
mediators initiating a vicious cycle of interconnected events- immune dysregulation,
cellular proliferation, disease perpetuation and tissue damage43 (Figure 6).
Inflammation results in the classical synovial pannus and bone marrow changes
consisting of cellular proliferation and new vessel formation. Cellular proliferation
includes sub-synovial areas of T and B cell aggregates resembling lymphoid tissue
capable of responding to local antigenic stimulation. Also present in the sub-synovial
area are macrophages, plasma cells and neutrophils. The synovium itself is
hypertrophied consisting of macrophage and fibroblast like synoviocytes. Cellular
communication is mediated by a host of cytokines, chemokines and growth factors.
43
Fig 6. Cytokine networks in RA. Following a yet unidentified stimulus recruitement and retention of
cellular and molecular components of the immune-inflammatory response. This is followed by
organisation of the local immune response and consequent on-going joint destruction: from Nature
clinical practice rheumatology; November 2005,Vol 1,No 1:McInnes and Liew.[with permission]
1.2.6 Immune Dysregulation
Dendritic cells are are found in the proliferating synovial tissue where they are able to
process and present antigens to T cells in synovial germinal centres and migrate to
central lymphoid organs where they activate naïve T cells. T cells then move back
into the synovium producing pro-inflammatory cytokines and driving B cell Ab
production. Dendritic cells may be stimulated by joint tissue debris such as
hyaluronan fragments, citrullinated synovial proteins, and type II collagen and
cartilage fragments. Dendritic cell functions are enhanced by pro-inflammatory
cytokines such as TNF, IL-1, and IL-6, while being inhibited by IL-10, TGF-β, and
44
prostaglandins. RA dendritic cells express fewer IL-10 receptors rendering them more
resistant to IL-10inhibition.2,44
Autoantibodies are one of the hallmark features of RA. De novo production of
pathological antibodies has been shown in RA synovial tissue. B and T cells are
found as aggregates in synovial tissues with some in highly developed germinal
centers known as tertiary lymphoid tissue (TLT). Histologic patterns vary from patient
to patient showing no correlation with disease activity, although areas of
granulomatous changes may possibly be associated with extra-articular disease.45
Tertiary lymphoid tissue is associated with many other inflammatory conditions
reflecting persistent immunological activation. Local secretion of lymphotoxin (LT-α,
LT-β) and chemokines [C-X-C motif (CXCL) 13, chemokine ligand (CCL21), CCL20
and CXCL12] modulates development of TLT. The majority of the cells is T cells (3050%) surrounding B cells and plasma cells, rich in IgM RF and ACPA IgG
antibodies.43,45 Synovial T cells are rich in CD28, an important co-stimulatory
molecule required for maximal response. A sub-population of synovial T cells lack
CD28 expression, acting as cytotoxic cells responding to auto-antigens. This may be
an important mechanism for treatment failures to biologic agents specifically targeting
T cell co-stimulation (Abatacept).45 IL-1 interacts synergistically with another costimulatory molecule CD40 binding to its ligand on T cells (CD40L) to produce GMCSF. Synovial T cells have high level of expression of receptors for CCR5 that
preferentially allow chemotaxis of Th1 T cells. People with a non-functional CCR5
genotype are protected from human immunodeficiency virus (HIV) infection and may
be at lower risk of RA. Production of IL-12 and IL-23 by dendritic cells favours Th1
and Th17 differentiation. Cytokine secretion is influenced by Th1 cells (IFN-γ, IL- 12,
IL-6, TNF and IL-10).46 T cell phenotypes in RA synovium include Th17 and Treg
cells. Th 17 cells secrete a potent pro-inflammatory cytokine IL-17. Tregs in RA have
a defect in the suppression of TNF and IFN- γ production by other T cells and
monocytes.47 APC, including B cells, activate T cells to initiate/perpetuate the local
immune reaction. B cell maturation within the TLT reflects a local antigen-driven
process.48 B cell-activating factor (BAFF) or B lymphocyte stimulator (BLyS) is a
45
cytokine essential for peripheral B cell survival with elevated serum levels seen in
patients with RA. BAFF is also produced by RASF. Cell contact with and production
of factors such as IL-6, IL-8 and GM-CSF by stromal cells found in the synovium and
bone marrow of patients with RA promotes B cell proliferation and antibody
production. The importance of B cells in RA pathogenesis is underscored by the
success of B cell depleting therapies (rituximab) in the management of refractory RA.
1.2.7 Cellular proliferation
Cellular recruitment, expression of adhesion molecules and angiogenesis are
mediated by a host of cellular and molecular components. Macrophages are key
producers of pro-inflammatory cytokines and chemokines in RA. Cytokines together
with growth factors (M-CSF, GM-CSF) regulate the balance between classical
inflammatory macrophages (M1) and those with an anti-inflammatory or tissue repair
phenotype (M2). Cytokines such as TNF are not only produced by macrophages,
which also respond to TNF in an autocrine manner, favouring further inflammatory
macrophage proliferation and differentiation of monocytes. This feedback loop is
likely to become entrenched in established RA and more difficult to manage, thereby
contributing to perpetuation of synovitis and chronicity.2 The extent of macrophage
synovial infiltration correlates with radiographic progression.49 Imbalances between
production of pro- and anti-inflammatory cytokines, such as increased production of
IL-1 and underproduction of IL-10, facilitate synovial tissue proliferation.50 Proinflammatory cytokines released by macrophages include TNF, IL-1, IL-12, IL-15, IL18, IL-23, IL-27, chemokines and growth factors. TNF is the principle cytokine,
increasing expression of other cytokines, prostaglandins (PGs) and adhesion
molecules. TNF also affects vascular permeability, allowing for movement of immune
complexes, complement, neutrophils and other cells into the synovium (Figure 7).
Cellular contact with fibroblasts promotes the release of IL-6, while adhesion
molecule expression increases under the influence of the pro-inflammatory cytokine
milieu.
46
Mast cells stimulate synovial cells to release PG E2 and collagenase, while heparin
released from these cells influences bone resorption. The proliferation of mast cells in
synovial tissue and fluid is related to the degree of lymphocyte infiltration with the
number of mast cells correlating with the clinical intensity of synovitis.51
Natural killer (NK) cells within the RA synovial infiltrate promote B cell production of
RFs and are also a source of serine proteases. Numerous CD56 positive natural killer
cells are found in RA synovium and result in increased pro-inflammatory cytokine
secretion by macrophages and T cells.
Neutrophils are found in abundance in RA synovial fluid, but sparsely in synovial
tissue. Neutrophils in synovial fluid amplify the inflammatory response by engaging
with immune complexes generating ROS and reactive nitrogen species (RNS), and
mobilization of neutrophil granules, resulting in release of proteases and cytokines
such as IL-1 and oncostatin M (member of the IL-6 familly). Neutrophil activation also
results in the formation of PGs and leukotrienes (LTs) from arachidonic acid released
from cell membrane phospholipids by phospholipase A2. The cytokines TNF and IL-1
induce the COX 2 gene, resulting in formation of pro-inflammatory PGs. LTs,
specifically LTB4 in the case of neutrophils, are produced from arachidonic acid via
the activity of 5’-lipoxygenase. LTB4 is a potent chemoattractant for neutrophils,
monocytes/macrophages and eosinophils. Activation of the 5’-lipoxygenase pathway
also results in formation of lipoxins from arachidonic acid; these have an antiinflammatory action, inhibiting neutrophil adhesion, chemotaxis and cytokine
production.
Mesenchymal cells in the bone marrow of RA patients express enhanced NFkB
signaling suggesting activation. These cells have the potential to differentiate into
fibroblast-like cells.52 Animal models have demonstrated movement of activated
mesenchymal marrow cells through cortical bone with consequent development of
synovitis under the modulation of cytokines.53
47
Fig 7. Cellular targets of TNF. TNF is released from cells as a soluble cytokine (sTNF, a homotrimer of
17-kDa monomers) after being enzymatically cleaved from its cell surface-bound precursor (a
homotrimer of 26-kDa monomers) by TNF-α-converting enzyme (TACE). Many immune and nonimmune cell types can produce TNF, including macrophages, T cells, mast cells, granulocytes, natural
killer (NK) cells, fibroblasts, neurons, keratinocytes and smooth muscle cells. TNF interacts with two
distinct receptors—TNF receptor 1 (TNFR1) (p55, CD120a) and TNFR2 (p75, CD120b)—on a wide
variety of cell types to mediate their biologic functions: from Trends in pharmacological sciences. AntiTNF therapy in the injured spinal cord: Volume 32, Issue 2, February 2011, Pages 107–115; Emanuela
Esposito,Salvatore Cuzzocrea, doi: 10.1016/j.tips.2010.11.009 [with permission]
1.2.8 Disease perpetuation
An altered fibroblast phenotype prevalent in RA synovial tissue confers an
autonomous, activated state and may be one of the major drivers of chronic
inflammation. Fibroblasts are cells of mesenchymal origin and are found at sites of
bony and cartilaginous destruction in the rheumatoid joint. RA synovial fibroblasts
secrete a host of pro-inflammatory cytokines, matrix degrading enzymes and factors
that promote angiogenesis. Fibroblasts do not normally express HLA class II
antigens, neither do they produce inflammatory cytokines. These immunological
48
changes are evident in RASF, even at the onset of clinical disease. The aetiology and
mechanisms of these changes are not known, but debris from joint damage appears
to promote RASF activation and ongoing disease. Cartilage damage releases
fragments such as fibronectin and vitronectin which also increases RASF activation
characterized by increased expression of adhesion molecules, resulting in increased
production of MMPs. Activation of RASF is also mediated by growth factors such as
fibroblast growth factor (FGF), TGF-β and platelet-derived growth factor (PDGF). The
altered RASF phenotype may also be related to epigenetic modifications with
hyperacetylation and DNA demethylation having been demonstrated in the RASF
genome activating pro-inflammatory gene transcription and production of factors
which promote fibroblast proliferation.1 RASF also contains unique miRNAs which are
also related to increased demethylation. Cellular debris such as RNA and DNA
fragments can bind to TLR expressed on RASF and promote activation thereof,
especially in the presence of pro-inflammatory cytokines such as IL-1 and TNF.
Cytokines such as TNF, IL-1, IL- 6, IL-17 and adipokines are also potent activators of
RASF’s. These events are summarized in Figure 8.
49
Fig 8. Roles of fibroblasts in RA. FLS play a critical part in many pathogenic events in the RA
synovium. They can contribute to pathology through a reduced ability to undergo apoptosis (forming
pannus), the production of proteases that degrade the extracellular matrix, and invasion into cartilage.
In addition, FLS produce a variety of molecules that modulate growth, inflammation, angiogenesis, and
cell recruitment, and induce activation of and cytokine production by immune cells. Abbreviations:
CCL2, CC-chemokine ligand 2; CXCL10, CXC-chemokine ligand 10; FLS, fibroblast-like synoviocytes;
GM-CSF, granulocyte-macrophage colony-stimulating factor; MMP, matrix metalloproteinase; PDGF,
platelet-derived growth factor; RA, rheumatoid arthritis; TGF-β, transforming growth factor β; VEGF,
vascular endothelial growth factor : from Nature Reviews Rheumatology.Duality of fibroblast-like
synoviocytes in RA: passive responders and imprinted aggressors: Nunzio Bottini and Gary S.
Firestein, 9, 24-33 (January 2013) doi:10.1038/nrrheum.2012.190) [with permission]
Heat shock protein (HSP) are stress response proteins released following tissue
damage related to physical stress, oxygen and nutrient deprivation, bacterial
infections and exposure to ROS/RNS. In a rat model of arthritis, T lymphocytes were
found to recognise mycobacterial HSP65 which was cross-reactive with cartilage
proteoglycan epitopes.54 Binding of HSPs to synovial TLR-4 and T cells can promote
disease perpetuation while certain HSPs stimulate rapid proliferation of synovial T
cells.55
50
The degradation of cartilage matrix components results in the formation of neoepitopes, potentially acting as auto-antigens. Immune complex deposition on articular
collagenous tissue serves as a strong and persistent stimulus for the generation of
chemo-attractants promoting sustained cellular infiltration of the synovial pannus. The
immune complexes in RA contain numerous proteins, including complement,
immunoglobulin, acute phase reactants, type II collagen, albumin, DNA and
fibrinogen.
1.2.9 Tissue damage
Osteoclasts are the primary effectors of bone resorption and are found in abundance
at the interface of proliferating synovium and bone. Mature osteoclasts are derived
from monocytes/macrophages requiring specific cytokine signals for maturation.
Osteoclasts secrete proteinases and possess a proton pump to allow for resorption of
the bony matrix and dissolution of calcium. Key proteinases are the MMPs and
cathepsins which remove non-mineralised areas of bone. The proton pump creates
an acidic environment between the osteoclast and bone surface. MMPs such as
MMP1, 2 and 9 are derived from synovial macrophages and mediate joint destruction
together with other MMPs, cathepsins, RANKL, angiogenic factors (VEGF), cytokines
(IL-1, IL-6) and chemokines (IL-8, MCP2) released from RASF.
Key molecular components necessary for osteoclast maturation include signals
derived from activated T cells and RA synovial fibroblasts. RANKL binds to its surface
receptor on osteoclasts, receptor activator for nuclear factor kappa B (RANK),
initiating differentiation, survival and activity.56 RANKL can also bind to a soluble
decoy receptor OPG that prevents RANKL/RANK interaction. Osteoclast formation is
also supported by TNF, IL-1 and IL-17 by promoting RANKL expression. TNF is
capable of directly binding to osteoclasts via its own receptor, thereby boosting
osteoclast formation and consequent bone resorption. Osteoclasts also mediate
resorption of the mineralized cartilage layer.57
51
In addition to bone loss at sites of the expanding synovial pannus and peri-articular
regions, a generalized decrease in bone mineral density is a well-recognized feature
of RA. Peri-articular bone loss is evident on bone mineral density (BMD) assessment
and is associated with joint damage and disease progression.58,59 Bone loss also
occurs at sites not distal to the synovium, thus local factors such as decreased
mobility and local bone marrow inflammation may also be etiological considerations.
Generalized osteopenia in RA is related to chronic inflammation and is associated
with disease activity.60,61 The pro-inflammatory circulating cytokine milieu is probably
also implicated in the pathogenic mechanisms inducing systemic bone loss.62
MMPs and aggrecanases are the the key mediators of cartilage destruction in RA.
Cartilage is composed of type II collagen (70%), proteoglycans, non-collagenous
matrix proteins (COMP) and chondrocytes. Proliferating synovial macrophages and
fibroblast-derived proteinases such as MMPs result in the degradation of collagen
and release of fragments carboxy-terminal collagen crosslinks (CTX)-II,63,64 and an
increase in cartilage turnover. Cartilage breakdown also occurs in areas not in
contact with synovium due to the dispersal of proteinases released into the synovial
fluid from neutrophils and other inflammatory cells in the synovial fluid.
Synovial pannus requires new vessel formation to proliferate such that ultrasound
Doppler activity in the RA synovium correlates with the future development of
erosions. Histological studies demonstrate proliferating vessels in inflamed joints
only.65 Vascular endothelial growth factor (VEGF) is the key growth factor initiating
new vessel formation. Pro-inflammatory cytokines such as IL-1, IL-6, TNF and TGF-β
stimulate the release of VEGF, resulting in endothelial cell proliferation. TNF may also
have a direct effect on endothelial cells promoting angiogenesis.66 The new vessels
in the RA synovium demonstrate abnormal architecture and are dysfunctional
resulting in ‘leakage’ of more inflammatory mediators into the synovium and
inadequate perfusion to synovial tissue.67
The interaction of genetic and environmental factors resulting in chronic selfperpetuating inflammation results in the clinical manifestations seen in RA. Recent
52
advances in the understanding of these pathogenic mechanisms have not only
resulted in novel approaches to the management of RA, but also shed light on
numerous other physiological and pathological processes such as aging, wound
healing, malignancies, autoimmune inflammatory disorders and ischeamic heart
disease (IHD).
1.3
Biomarkers in RA
With the recent advances in the management of patients with RA, the need for early
diagnosis and rapid disease control is paramount to limit morbidity and premature
mortality from RA. Particularly in resource poor settings, prioritization of services to
allow early diagnosis, and prognostication, as well as the need to institute appropriate
cost effective therapy makes the prospect of personalized medicine all the more
critical. Clinical and imaging strategies, as well as laboratory biomarkers are being
used to diagnose and monitor disease progression, usually as composite scores.
However, current biomarkers in use, although valuable, fail to diagnose or monitor
disease activity effectively. Measures associated with a good clinical response may
not be able to identify patients with ongoing joint damage, hence the importance of
identifying novel markers able to bridge the disconnect between clinical response and
joint damage. Candidate novel markers that could contribute to improving RA
management are those integrally involved in the immunopathogenesis of RA. Ideal
biomarkers should facilitate diagnosis, characterize severity, and predict response to
therapy, morbidity and premature mortality.
1.3.1 Clinical measures and acute phase response measures
Clinical measures such as joint counts for swelling or tenderness and global
assessments of disease activity by the physician and patient using a visual analog
scale are commonly used to assess disease activity and response to therapy. Clinical
and demographic features associated with a response to methotrexate therapy
include early disease, low functional impairment, male sex, young age, elderly onset
RA, low disease activity and non-smoker status.68,69 However, studies have reported
53
conflicting reports with data from the Norfolk Arthritis Register failing to show age,
age at onset, or gender to be predictive of response to MTX.70 In the Swedish
Pharmacotherapy Trial (SWEFOT), a poor response to MTX was associated with
short duration of symptoms, younger age, female gender and current smoking
status.71,72 The erythrocyte sedimentation rate and C-reactive protein (CRP) are
included in some of the disease activity scores, but may be normal in as many as
30% of patients despite active disease. Unfortunately, even in patients with low
disease activity scores, ongoing radiographic damage and disease progression
occurs.
1.3.2 Imaging markers
Plain X-rays of the hands and feet are used as surrogate markers of disease
progression with radiographic scoring taking into account degree of bony destruction
and joint space narrowing as representing cartilage loss. Two popular scoring
measures are the Larsen and Sharp van der Heide methods, but these are used
mostly in clinical trials. The simplified erosion and narrowing score (SENS), a
modification of the Sharp van der Heide score may be more practical for clinical use.
However, radiographic changes are not commonly present in early disease, and
hence are no longer listed in the new RA criteria. Newer imaging modalities such as
ultrasound and magnetic resonance imaging (MRI) hold promise to monitor disease
progression and response to therapy early in disease management. Doppler
ultrasound activity and MRI marrow edema have been associated with erosive
radiographic progression. Ultrasound measures of synovitis have been shown to be
useful in monitoring response to MTX and are now being used as an outcome
measure in some clinical trials.73-75 The utility of newer imaging modalities as
biomarkers in RA still requires validation with cost and expertise being significant
encumbrances.
54
1.3.3 Genotyping
Meyer et al in a cohort of predominantly black South African RA patients, showed an
association of the SE with RA in 88 % of patients similar to other studies in
Caucasians, but the association did not add more predictive value than the presence
of ACPA, thus limiting clinical utility of shared epitope genotyping.26 The presence of
the SE may, however, have prognostic value as several studies have shown an
association with more aggressive disease.76-78 HLA DRB1*04 together with ACPA
status is associated with resistance to disease modifying anti-rheumatic drug
(DMARD) therapy,79 SE-positive patients may benefit from combination DMARDs
from the outset as opposed to SE-negative patients.80 Homozygous SE patients have
a 2-fold increase in cardiovascular (CV) mortality particularly in the case of the HLADRB1*01/*04 genotypes.81-84
Other genetic loci implicated in predisposition for developing RA include PTPN 22,
TRAF1/C5 and STAT 4. The PTPN 22 risk allele codes for lymphoid tyrosine
phosphatase that is involved in inhibition of T cell activation. The TRAF1/C5 risk allele
locus is related to the genes encoding complement C5 and TNF receptor-associated
factor 1. STAT 4 is associated with transcription of genes encoding interferon-α.85-87
Genetic polymorphisms of MMP 3 have been shown to be associated with increased
joint destruction in RA.63,88
Genetic polymorphisms of purine and pyrimidine metabolism may also influence the
response to MTX and several genetic variants have the potential to act as biomarkers
of response to MTX.89,90 Gene expression in the synovia and peripheral blood
mononuclear cells (PBMC) also varies with MTX treatment. A decrease has been
noted in some RA related genes in synovial fibroblasts and an increase in IL-4 and IL10 gene expression in PBMC in RA patients after treatment with MTX.91 Genetic
expression of CCL4, IL-8 and IL-1β may be predictive of response to TNF
blockade.92-95
55
Abnormal expression of miRNAs in RA synovia has been documented and miR-16,132,-146 and -155 identified as potential biomarkers in RA. Circulating levels of miR132 were found to be lower in RA patients compared to healthy controls, while levels
of miR-16 were found to correlate with disease activity.96-98 Elevated levels of miR125b may be predictive of a good response to.B cell depletion therapy with
Rituximab.
The routine use of genotyping is prohibitive as access to testing is often only
available in specialised laboratories, sometimes requiring synovial tissue,
necessitating a biopsy. These factors also make the prospect of large studies to
validate the role of genotyping more difficult.
1.3.4 Autoantibodies
Rheumatoid factor (RF) is one of the well-established laboratory markers associated
with RA, but sensitivity and specificity, especially in early disease may be as low as
50%. Numerous conditions are associated with a false-positive test (infections,
malignancies and connective tissue disorders). Pre-clinical detection of high levels of
RF is, however, a risk factor for the development of RA.99,100 The evaluation of RF
isotypes increases specificity with the presence of elevated IgM and IgA RFs very
suggestive of RA. High levels are usually seen in patients with severe disease, but
despite the relationship with RA, changes in the level of RF with treatment have been
found not to be clinically useful to monitor disease activity or response to therapy.101
Studies of RF factor levels as predictors of response to TNF blocker therapy have
reported varying results with some showing no predictive power102-104 and others
predicting a negative response, especially in the presence of high levels of IgA RF.105107
High serum levels of RF may be predictive of a more favourable response to
rituximab therapy.108,109
Cardiovascular co-morbidity is increased in patients who are RF seropositive, with RF
reported to be associated with left ventricular (LV) dysfunction and an independent
56
risk factor for IHD. RF is also associated with an increased all-cause mortality,
unrelated to RA disease activity.110-113
The absolute levels of RF and ACPA, if increased greater three fold above normal
increases the likelihood of diagnosing RA114 (Table 1). The addition of the ACPA to
RF increases specificity of diagnosing RA to greater than 90%, but sensitivity is still
around 50-60% especially in early disease. Antibody positivity also has prognostic
value with ACPA being associated with erosive disease and radiographic
progression.115-119 Erosive disease occurs earlier and with increased frequency in
patients with ACPA positivity.120 The combined presence of RF and SE has an
interactive effect on ACPA status with respect to a poor prognosis,115 with the greater
spectrum of ACPA isotypes present associated with more severe radiographic
changes.116
Studies of antibody status as predictors of response to TNF blockers have produced
conflicting data,105,121 however, sero-positivity for RF and/or ACPA and elevated
immunoglobulins (IgG) has been shown to be associated with a better response to
the B cell-depleting agent rituximab.122,123 A decrease in ACPA titres with biologic
DMARDs has correlated with clinical response and may be a marker of response to
therapy,107,124-132 but results from other studies have shown no such correlation.133-136
Higher levels may be associated with a poor response to TNF antagonism and B celldepletion therapy with rituximab.133,137 The clinical utility of ACPA to monitor disease
progression has not yet been established. ACPA-positive patients have greater
frequencies of subclinical atherosclerotic disease being independently associated
with future IHD and associated LV dysfunction.111,138,139
More recently a new antibody system related to, but independent of ACPA has been
characterized, namely anti-carbamylated protein (anti-CarP). Carbamylation involves
the post-translational modification of lysine to homocitrulline as opposed to
conversion of arginine to citrulline for ACPA (Figure 9). This reaction occurs during
inflammation following release of myeloperoxidase from neutrophils140-142 the
57
presence of anti CarP antibodies in RA was found to be associated with more severe
disease/joint damage in a group of ACPA-negative patients.140
Fig 9. Illustration of citrullination and carbamylation. Citrullination (A) and carbamylation (B) occur on
different amino acids via different mechanisms, but yield similar end-products: from PNAS 2011 108
(42) 17372-17377 ©2011 by National Academy of Sciences (open access)
1.3.5 Novel laboratory markers
Cytokines are important molecular components of the immune-inflammatory
response and are integrally involved in the pathogenesis of RA. Their importance is
underscored by the success of numerous innovative therapeutic agents targeting
specific cytokines or their biologic pathways (Figure 10). Measurement of circulating
levels of cytokines, chemokines, certain growth factors and degradation products
associated with RA pathogenesis is being explored in the search for cost-effective,
reliable biomarkers.
58
Fig 10 Pathogenesis of RA.
APCs present self or nonself antigens to T cells, which differentiate into TH1 or TH17 cells that induce
macrophages to secrete proinflammatory cytokines and help B cells to produce (auto)antibodies that
bind to target antigens forming immune complexes. These immune complexes can bind complement
and engage complement receptors and/or Fc receptors on macrophages, augmenting secretion of
TNF, IL-1 and IL-6. These cytokines lead to the clinical and laboratory manifestations of RA, as well as
cartilage and bone damage, by virtue of chondrocyte and osteoclast activation (the latter primarily via
the RANKL–RANK system). These cytokines can also alter and activate differentiation and function of
T cells and B cells operating upstream in RA pathogenesis. TREG cells can inhibit the function of TH1
and TH17 cells in a physiologic environment, but are inhibited by cytokines produced in the
inflammatory response. Abbreviations: APC, antigen-presenting cell; RA, rheumatoid arthritis; RANK,
receptor activator of nuclear factor-κB; RANKL, RANK ligand; TCR, T-cell receptor; TGF-β,
transforming growth factor β; TH, T-helper cell; TLR, Toll-like receptor; TREG, T-regulatory cell: from
Nature Reviews Rheumatology, 8, 235-243 (April 2012) The pathogenesis of rheumatoid arthritis: new
insights from old clinical data? Josef S. Smolen, Daniel Aletaha and Kurt Redlich [with permission].
59
Even in early RA local and systemic bone loss is associated with inflammation and
patient functional status.143 Radiographic erosive disease has been found to be
associated with decreased BMD,59,144,145 while serum TNF levels have been found to
be inversely correlated to BMD.146 RANKL and OPG play an important role in bone
resorption and formation (chapter 2). The ratio of RANKL/OPG may have diagnostic
value as higher levels are found in RA compared to healthy controls with serum OPG
correlating negatively with disease activity and RANKL correlating positively with
CRP.147 A high baseline RANKL/OPG ratio is strongly predictive of yearly
radiographic progression,148 while a low serum RANKL level at baseline was found to
be an independent predictor of remission.69 A decrease in the RANKL/OPG ratio was
associated with remission in patients treated with IL-6 antagonists, resulting from an
increase in OPG and bone formation.147,149 B cell-depletion therapy is also associated
with a lower RANKL/OPG ratio. Patients treated with TNF blockers experience a
decrease in serum RANKL levels in association with a decrease in disease activity,150
thus RANKL/OPG may be of potential value in monitoring response to therapy.
Baseline RANKL levels and serum RANKL/OPG ratio may also identify a subset of
patients more likely to respond to TNF treatment.
Biomarkers reflecting enzymatic processes associated with breakdown of bone and
cartilage have been shown to be associated with radiographic progression.
Independent predictors of radiographic outcome include MMP3 and COMP.151 MMP3
has also been found to correlate with erosive disease despite a normal CRP with
some studies showing no predictive potential regarding radiographic progression
independent of RF or ACPA status.151-154 Combining disease activity score (DAS) 28
and MMP3 was demonstrated to be better than other outcome measures in RA for
monitoring response to therapy.155 The post-therapy normalization of MMP3 levels
with IL-6 blockade which tocilizumab was predictive of sustained remission.153 MMP3
levels at baseline and decrements thereof correlated with response to combination
therapy with infliximab and MTX.156
60
COMP maintains the structural integrity of cartilage and is a measure of cartilage
turnover. In an early RA cohort changes in circulating COMP levels were associated
with radiologic outcome, with low serum levels of COMP at baseline being predictive
of a rapid ACR70 response to adalimumab.157 YKL-40 is a cartilage glycoprotein
secreted by chondrocytes and synovial tissue in the presence of joint inflammation
which decreases in response to treatment. YKL-40 has been included in a multibiomarker score assessing disease activity in RA.158-160
Breakdown products from collagen and non-collagenous components of bone and
cartilage (CTX-I and CTX-II) have also been shown to be associated with increased
risk of radiographic progression.161,162 Serum levels of cathepsin K have also been
shown to correlate with radiological damage in patients with RA. Cathepsin K plays
an important role in resorption of non-mineralized bone and a decrease in cathepsin
K-mediated bone resorption following biological therapy with IL-6 blockade has been
associated with a decrease in CTX I levels.163 Elevated urinary CTX II levels have
been found to be associated with rapid radiographic progression in patients with
established disease.164
Clusters of cytokines/chemokines, including TNF, IL-1, IL-6, IL-13, IL- 8, as well as
CCL2, CXCL10, CCL11 may have diagnostic value because they are elevated in
patients with RA when compared to healthy controls.165 Circulating
cytokine/chemokine profiles associated with disease severity include elevated CCL4,
IL-8, IL-2, IL-12, IL-17, and elevated IL-10 associated with mild disease.166 Lower IL4, IL-6, IFN-γ, and GM-CSF and IL-4 levels have been reported in patients with more
severe disease.166 Cytokine levels associated with disease activity include IL-1β, IL-6,
IL -18 and TNF,160,167,168 but other studies have shown no correlation between
cytokine levels and clinical measures of disease activity.92
Alterations in circulating cytokine levels, may, however, have the potential to monitor
response to therapy, with IL-7 levels decreasing in patients responding to MTX169 and
levels of VEGF declining following TNFα blockade.170 Radiographic progression has
been shown to be associated with alterations in mean circulating IL-6 levels over 2
61
years.160 The absolute plasma levels of IL-6 (>4.03 pg/ml) after MTX therapy may be
associated with radiographic progression,171 while IL-7 levels correlate with erosive
disease.156 An increase in IL-8 levels from baseline was found to correlate with
radiographic progression in patients on MTX monotherapy.172
Measures of the ratio of IL-1Ra/IL-1 produced by PBMC, as well as serum TNF levels
have the potential to predict response to therapy with MTX.68,91,173 High circulating
chemokine levels (CCL2 and CXCL8) at baseline have also been reported to be
associated with response to IL-6 inhibition,174 with low baseline levels of IL-6 being
associated with a more favourable response to IL-6 blockade.167 High levels of CCL2
and epidermal growth factor (EGF) were found to be predictive of response to
etenercept,175 with low levels of IL-2 predictive of remission in RA.69 Some biological
therapies such as B cell-depletion are given at 6 to 9 monthly intervals and early
predictors of response could add value to RA management. Decreases in CRP, in the
setting of higher CCL2 and EGF at 3 months after a dose of rituximab may be
predictive of responders.176 Higher baseline values of IL-6 were also associated with
a poor response to rituximab, with a significant reduction in circulating IL-6 levels
noted in responders.177 Patients with very high serum TNF values were found to be
more refractory to treatment.177
Measurement of growth factors associated with neovascularisation such as VEGF
has demonstrated higher levels in patients with RA, especially in early disease before
therapy. Levels of VEGF correlate with disease activity and decrease in response to
therapy in some studies.178-180
Co-morbidities such as accelerated atherosclerosis are an important cause of
premature mortality in patients with RA with cytokines seeming to be key pathogenic
mediators (Figure 11). Measures of endothelial dysfunction such as increased
expression of vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion
molecule-1 and endothelial leukocyte adhesion molecule-1 have also been
described in patients with RA,181 with VCAM-1 associated with increased common
carotid artery intima medialthickness (IMT) and plaque formation in RA patients.182
62
Cytokine levels, particularly ratios of pro-inflammatory to anti-inflammatory cytokines
such as the ratios of IL-1/IL-10 and IL-6/IL-10 have also been associated with new
coronary events in patients (non-RA) with previous non ST segment elevation acute
coronary syndrome (NSTEACS).183 Levels of IL-6 and TNF have been shown to be
associated with increased coronary arterial calcification in RA, with increased arterial
stiffness correlating with IL-6 levels.184,185
63
Fig 11. Schematic representation of T lymphocyte subset activation within atherosclerotic plaque.
Different self-structures are expressed in the intima of the vessel wall and subjected to
enzymatic/oxidative modifications. These modified molecules are taken up by dendritic cells and
presented to CD4+ T helper cells via MHC class II molecules. Antigen presentation promotes T
lymphocyte activation and secretion of different cytokines. Effector CD4+ Th1 cells produce proinflammatory cytokines, particularly IFN-γ, with proatherogenic effects. Activated Th2 lymphocytes
secrete IL-4 and IL-5 which promote the differentiation of B cells in plasma cells and the production of
specific antibodies. Some infiltrated T cells upon activation differentiate in Th17 lymphocytes
characterized by the production of IL-17, but their role remains controversial. Regulatory T cells
(Tregs) downregulate inflammation by secretion of TGF-β and IL-10. The pro-inflammatory mediators
released by activated T cells reduce the stability of the lesion promoting plaque rupture and thrombotic
events:
from The ScientificWorld Journal Volume 2012, Article ID 157534. T Lymphocyte Autoreactivity in
InflammatoryMechanisms Regulating Atherosclerosis. Elisabetta Profumo, Brigitta Buttari, Luciano
Saso, Raffaele Capoano,Bruno Salvati, and Rachele Rigan`o [open access]
64
Although correlations of these novel biomarkers with disease activity or as predictors
of response to treatment have been inconsistent and of limited clinical use as
independent markers, some of these novel biomarkers have been incorporated into
composite scores with other measures of disease activity, with the biomarker
component now commercially available (Figure 12). The composite score has been
found to be associated with disease activity, radiographic progression and ability to
discriminate responders from non-responders to MTX and TNF blockers.148,187 The
clinical utility of the multi-biomarker score appears promising but needs to be
validated in larger clinical trials. In a recent study of an early RA cohort, the pretreatment multi biomarker score was shown to be an independent predictor of
radiographic progression at 1 year.188
Fig 12. The Venn diagram lists the MBDA score biomarkers used to predict each MBDA score
component. This algorithm provides an MBDA score between 1 and 100 :from Ann Rheum Dis
2012;71:1692–1697. doi:10.1136/annrheumdis-2011-200963. [open access]
65
1.3.6 Comment
As the etio-pathogenic mechanisms underlying RA unfold more candidate biomarkers
and potential therapeutic targets are being identified. Recent advances in RA
management have brought with them challenges in respect of selection of specific
therapies for specific situations, although no single measure is likely to emerge as the
gold standard. Composite scores using clinical, imaging and laboratory measures are
likely to come closest to meeting the ideals of cost effective-personalized medicine.
66
Table 1: ACR/EULAR 2010 classification criteria for RA.49
To be applied to patients: (1) who have ≥ 1 joint with definite synovitis, excluding the DIP
joints, first MTP joints, and first CMC joints, and (2) in whom the synovitis cannot be
explained by another disease.
Criteria
A.
B.
C.
D.
Score
Joint involvement:
1 large joint
0
2 – 10 large jointsa
1
1 – 3 small joints (with or without involvement of large joints)
2
4 – 10 small jointsb (with or without involvement of large joints)
3
>10 joints (at least1 small joint)
5
Serology (at least 1 test result is needed for classification):
Negative RF and negative anti-CCP antibodies
0
Low-positive RF or low-positive anti-CCP antibodiesc
2
High-positive RF or high-positive anti-CCP antibodiesd
3
Acute phase reactants:
Normal CRP level and normal ESR
0
Abnormal CRP level or abnormal ESR
1
Duration of symptoms:
< 6 weeks
0
≥ 6 weeks
1
ACR, American College of Rheumatology; EULAR, European League Against Rheumatism; RA, rheumatoid
arthritis; DIP, distal interphalangeal; MTP, metatarsophalangeal; CMC, carpometacarpal; MCP,
metacarpophalangeal, PIP, proximal interphalangeal; IP, interphalangeal; RF, rheumatoid factor; anti-CCP, anticyclic citrullinated protein; CRP, C-reactive protein; ESR, erythrocyte sedimentation rate.
a
Large joints = shoulders, elbows, hips, knees, ankles.
Small joints = MCPs, PIPs, second – fifth MTPs, thumb IPs, wrists.
c
Low-positive is ≤ 3 times the upper limit ofnormal.
d
High-positive is > 3 times the upper limit of normal.
b
67
1.4
Rheumatoid Arthritis in the developing world
A paucity of data exists on the epidemiology of RA in developing countries with a
presumptive prevalence ranging from 0.5-1%.189 There is a perception that the
prevalence of RA is increasing in developing countries, but decreasing with a shift to
a more elderly onset of disease in developed regions such as Europe and
America.190,191 Most studies in developing areas reflect an increased prevalence in
urban areas in comparison to rural areas.189 There are numerous challenges peculiar
to the developing world including burden of communicable diseases, poor
socioeconomic factors and limited resources making the management of RA more
difficult.192 A West African study seems to suggest that RA may be milder in Africa in
comparison to a British cohort but more recent data from Africa report a disease
pheno-typically similar to that seen in the developing world.193-195 Genetic
susceptibility to RA may vary geographically with a lower frequency of the RA
associated risk allelle HLA-DR4 reported in Nigerians compared to South
Africans.196,197 In developing countries the higher prevalence of infections such as
tuberculosis (TB), HIV and viral hepatitis poses additional threats to health care.
There is a greater than 8-fold increased risk for TB due to the disease and its
treatment.192 Many people in the developing world are involved in manual labour and
a study in Chile noted that most manual workers with RA stopped active work 2 years
from disease diagnosis.198 Synthetic DMARD’s are the anchor drugs used to manage
the disease with only few patients having access to expensive biological therapies.
Synthetic DMARDs can have remarkable efficacy if started in early disease.
However, studies have highlighted the all too frequent delay in the diagnosis and
institution of appropriate management of RA in the developing world, with a
consequent negative impact on quality of life, functional status, prognosis and burden
on society.192,195
Early diagnosis and initiation of therapy are key factors that will ultimately result in
cost effective outcomes for all patients with RA, especially of importance in
developing countries. There is also very limited data available in Africa and the rest of
68
the developing world on clinical and laboratory biomarkers of disease severity and
response to therapy. More studies are needed that would contribute to estimation of
the burden of disease, identify markers of disease patterns and response to therapy.
This, in turn, would allow for better utilization of scarce resources ultimately
benefitting both patients with RA and Society at large.
To this end a collaborative effort involving two public sector rheumatology units in the
Gauteng province of South Africa recruited 171 DMARD-naïve early RA patients and
conducted a 2 year prospective study, the Gauteng Rheumatoid Evaluation
Assessment Trial (GREAT).
Objectives of the study included; i) characterization of disease burden with respect to
both physical and psycho-social aspects; ii) assessing response to therapy in routine
care; iii) characterization of auto-antibody, cytokines/chemokines, MMP-3, cartilage
metabolites and genotype profiles associated with disease activity and response to
therapy; and iv) investigation of clinical, radiographic and serological measures
associated with radiographic damage and progression.
GREAT is a large and ongoing study encompassing many aspects of RA in the study
population that has led to the completion of two theses (Dr PWA Meyer and Dr B
Hodkinson) focused on research areas distinct from those presented in this thesis.
Abstracts of published articles are listed in the appendix and references listed at the
end of each chapter.
69
References
1.
Neumann E, Lefèvre S, Zimmermann B, Gay S, Müller-Ladner U. Rheumatoid
arthritis progression mediated by activated synovial fibroblasts. Trends in
Molecular Medicine 2010; 16(10):458-468.
2.
Lutzky V, Hannawi S, Thomas R. Cells of the synovium in rheumatoid arthritis.
Dendritic cells. Arthritis Research & Therapy 2007; 9(4):219.
3.
Strietholt S, Maurer B, Peters MA, Pap T, Gay S. Epigenetic modifications in
rheumatoid arthritis. Arthritis Research & Therapy 2008, 10(5):219.
4.
Dinarello CA. Historical insights into cytokines. European Journal of
Immunology 2007; 37(Suppl 1):S34-S45.
5.
Newton K, Dixit VM. Signaling in innate immunity and inflammation. Cold
Spring Harbor Perspectives in Biology 2012; 4(3):a006049.
6.
So A, Busso N. The concept of the inflammasome and its rheumatologic
implications. Joint Bone Spine 2014; 81(5):398-402.
7.
Dinarello CA. Interleukin-1 in the pathogenesis and treatment of inflammatory
diseases. Blood 2011; 117(14):3720-3732.
8.
Dinarello CA. Overview of the interleukin-1 family of ligands and receptors.
Seminars in Immunology 2013; 25(6):389-93.
9.
Akdis M, Burgler S, Crameri R, Eiwegger T, Fujita H, Gomez E, Klunker S,
Meyer N, O’Mahony L, Palomares O, Rhyner C, Ouaked N, Schaffartzik A,
Van De Veen W, Zeller S, Zimmermann M, Akdis CA. Interleukins, from 1 to
37, and interferon-γ: receptors, functions, and roles in diseases. Journal of
Allergy and Clinical Immunology 2011; 127(3):701-721.
10.
Jazayeri JA, Carroll GJ, Vernallis AB. Interleukin-6 subfamily cytokines and
rheumatoid arthritis: role of antagonists. International Immunopharmacology
2010; 10(1):1-8.
11.
Niu N, Qin X. New insights into IL-7 signaling pathways during early and late T
cell development. Cellular & Molecular Immunology 2013; 10(3):187-189.
70
12.
Mathew SJ, Haubert D, Krönke M, Leptin M. Looking beyond death: a
morphogenetic role for the TNF signalling pathway. Journal of Cell Science
2009; 122(Pt 12):1939-1946.
13.
Yamana J, Yamamura M, Okamoto A, Aita T, Iwahashi M, Sunahori K, Makino
H. Resistance to 1L-10 inhibition of interferon gamma production and
expression of suppressor of cytokine signaling 1 in CD4+ T cells from patients
with rheumatoid arthritis. Arthritis Research & Therapy 2004; 6(6):R567-R577.
14.
Gordon KJ, Blobe GC. Role of transforming growth factor-beta superfamily
signaling pathways in human disease. Biochimica et Biophysica Acta 2008;
1782(4):197 – 228.
15.
Legge KL, Gregg RK, Maldonado-Lopez R, Li L, Caprio JC, Moser M,
Zaghouani H. On the role of dendritic cells in peripheral T cell tolerance and
modulation of autoimmunity. The Journal of Experimental Medicine 2002;
196(2):217-227.
16.
Warrington R, Watson W, Kim HL, Antonetti FR. An introduction to
immunology and immunopathology. Allergy, Asthma & Clinical Immunology
2011; 7 (Suppl 1):S1.
17.
Thomas R. Dendritic cells and the promise of antigen-sepcific therapy in
rheumatoid arthritis. Arthritis Research & Therapy 2013; 15(1):204.
18.
Li J, Hsu HC, Mountz JD. Managing macrophages in rheumatoid arthritis by
reform or removal. Current Rheumatology Reports 2012; 14(5):445-454.
19.
Arazna M, Pruchniak MP, Demkow U. Neutrophil extracellular traps in bacterial
infections: strategies for escaping from killing. Respiratory Physiology &
Neurobiology 2013; 187(1):74-77.
20.
Carroll MV, Sim RB. Complement in health and disease. Advanced Drug
Delivery Reviews 2011; 63(12):965-975.
21.
Lundy SK, Sarkar S, Tesmer LA, Fox DA. Cells of the synovium in rheumatoid
arthritis. T lymphocytes. Arthritis Research & Therapy 2007; 9(1):202.
71
22.
Blüml S, McKeever K, Ettinger R, Smolen J, Herbst R. B-cell targeted
therapeutics in clinical development. Arthritis Research & Therapy 2013;
15(Suppl 1):S4.
23.
Klinker MW, Lundy SK. Multiple mechanisms of immune suppression by B
lymphocytes. Molecular Medicine 2012; 18:123-137.
24.
Bryant VL, Ma CS, Avery DT et al. Cytokine-mediated regulation of human B
cell differentiation into Ig-secreting cells: predominant role of IL-21 produced
by CXCR5+ T follicular helper cells. The Journal of Immunology 2007;
179(12):180-190.
25.
Kurkó J, Besenyei T, Laki J, Glant TT, Mikecz K, Szekanecz Z. Genetics of
rheumatoid arthritis - a comprehensive review. Clinical Reviews in Allergy &
Immunology 2013; 45(2):170-179.
26.
Meyer PW, Hodkinson B, Ally M, Musenge E, Wadee AA, Fickl H, Tikly M,
Anderson R. HLA-DRB1 shared epitope genotyping using the revised
classification and its association with circulating autoantibodies, acute phase
reactants, cytokines and clinical indices of disease activity in a cohort of South
African rheumatoid arthritis patients. Arthritis Research & Therapy 2011;
13(5):R160.
27.
Deane KD, El-Gabalawy H. Pathogenesis and prevention of rheumatic
disease: focus on preclinical RA and SLE. Nature Reviews Rheumatology
2014; 10(4):212-228.
28.
Lee DM, Phillips R, Hagan EM, Chibnik LB, Costenbader KH, Schur PH.
Quantifying anti-cyclic citrullinated peptide titres: clinical utility and association
with tobacco exposure in patients with rheumatoid arthritis. Annals of the
Rheumatic Diseases 2009; 68(2):201-208.
29.
Niedermeier M, Pap T, Korb A. Therapeutic opportunities in fibroblasts in
inflammatory arthritis. Best Practice & Research Clinical Rheumatology 2010;
24(4):527-554.
30.
Combe B. Progression in early rheumatoid arthritis. Best Practice & Research
Clinical Rheumatology 2009(1); 23:59-69.
72
31.
MacGregor AJ, Snieder H, Rigby AS, Koskenvuo M, Kaprio J, Aho K, Silman
AJ. Characterizing the quantitative genetic contribution to rheumatoid arthritis
using data from twins. Arthritis & Rheumatism 2000; 43(1):30-37.
32.
Carrier N, Cossette P, Daniel C, de Brum-Fernandes A, Liang P, Ménard HA,
Boire G. The DERAA HLA-DR alleles in patients with early polyarthritis:
protection against severe disease and lack of association with rheumatoid
arthritis autoantibodies. Arthritis and Rheumatism 2009; 60(3):698-707.
33.
Gregersen PK, Silver J, Winchster RJ. The shared epitope hypothesis. An
approach to understanding the molecular genetics of susceptibility to
rheumatoid arthritis. Arthritis and Rheumatism 1987; 30(11):1205-1213.
34.
du Montcel ST, Michou L, Petit-Teixeira E, Osorio J, Lemaire I, Lasbleiz S,
Pierlot C, Quillet P, Bardin T, Prum B, Cornelis F, Clerget-Darpoux F. New
classification of HLA-DRB1 alleles supports the shared epitope hypothesis of
rheumatoid arthritis susceptibility. Arthritis and Rheumatism 2005; 52(4):10631068.
35.
Michou L, Croiseau P, Petit-Teixeira E, du Montcel ST, Lemaire I, Pierlot C,
Osorio J, Friqui W, Lasbleiz S, Quillet P, Bardin T, Prum B, Clerget-Darpoux F,
Cornélis F; European Consortium on Rheumatoid Arthritis Families. Validation
of the reshaped shared epitope HLA-DRB1 classification in rheumatoid
arthritis. Arthritis Research & Therapy 2006; 8(3):R79.
36.
Klein K, Ospelt C, Gay S. Epigenetic contributions in the development of
rheumatoid arthritis. Arthritis Research & Therapy 2012; 14(6):227.
37.
Ceribelli A, Nahid MA, Satoh M, Chan EK. MicroRNAs in rheumatoid arthritis.
FEBS Letters 2011; 585(23):3667-3674.
38.
Liao KP, Alfredsson L, Karlson EW. Environmental influences on risk for
rheumatoid arthritis. Current Opinion in Rheumatology 2009; 21(3):279-283.
39.
Hughes LB, Morrison D, Kelley JM, Padilla MA, Vaughan LK, Westfall AO,
Dwivedi H, Mikuls TR, Holers VM, Parrish LA, Alarcón GS, Conn DL, Jonas
BL, Callahan LF, Smith EA, Gilkeson GS, Howard G, Moreland LW, Patterson
N, Reich, D, Bridges SL Jr. The HLA-DRB1 shared epitope is associated with
73
susceptibility to rheumatoid arthritis in African Americans through European
genetic admixture. Arthritis and Rheumatism 2008; 58(2):349-358.
40.
Mikuls TR, Payne JB, Yu F, Thiele GM, Reynolds RJ, Cannon GW, Markt J,
McGowan D, Kerr GS, Redman RS, Reimold A, Griffiths G, Beatty M,
Gonzalez SM, Bergman DA, Hamilton BC 3rd, Erickson AR, Sokolove J,
Robinson WH, Walker C, Chandad F, O’Dell JR. Periodontitis and
Porphyromonas gingivalis in patients with rheumatoid arthritis. Arthritis &
Rheumatology 2014; 66(5):1090-1100.
41.
Cole BC, Griffiths MM. Triggering and exacerbation of autoimmune arthritis by
the Mycoplasma arthritidis superantigen MAM. Arthritis and Rheumatism 1993;
36(7):994-1002.
42.
Saal JG, Steidle M, Einsele H, Müller CA, Fritz P, Zacher J. Persistence of B19
parvovirus in synovial membranes of patients with rheumatoid arthritis.
Rheumatology International 1992; 12(4):147-151.
43.
McInnes IB, Liew FY. Cytokine networks - towards new therapies for
rheumatoid arthritis. Nature Clinical Practice Rheumatology 2005; 1:31-39.
44.
MacDonald KP, Pettit AR, Quinn C, Thomas GJ, Thomas R. Resistance of
rheumatoid synovial dendritic cells to the immunosuppressive effects of IL-10.
Journal of Immunology 1999; 163(10):5599-5607.
45.
Warrington KJ, Takemura S, Goronzy JJ, Weyand CM. CD4+, CD28- T cells in
rheumatoid arthritis patients combine features of the innate and adaptive
immune systems. Arthritis and Rheumatism 2001; 44(1):13-20.
46.
Nagata S, Kawane K. Autoinflammation by endogenous DNA. Advances in
Immunology 2011; 110:139-161.
47.
Nagata S. Rheumatoid polyarthritis caused by a defect in DNA degradation.
Cytokine & Growth Factor Reviews 2008; 19(3-4):295-302.
48.
Kim HJ, Berek C. B cells in rheumatoid arthritis. Arthritis Research 2000;
2(2):126-131.
74
49.
Finnegan A, Ashaye S, Hamel KM. B effector cells in rheumatoid arthritis and
experimental arthritis. Autoimmunity 2012; 45(5):353-363.
50.
Müller-Ladner U, Roberts CR, Franklin BN, Gay RE, Robbins PD, Evans CH,
Gay S. Human IL-1Ra gene transfer into human synovial fibroblasts is
chondroprotective. Journal of Immunology 1997; 158(7):3492-3498.
51.
Malone DG, Wilder RL, Saavedra-Delgado AM, Metcalfe DD. Mast cell
numbers in rheumatoid synovial tissues. Correlations with quantitative
measures of lymphocytic infiltration and modulation by anti-inflammatory
therapy. Arthritis and Rheumatism 1987; 30(2):130-137.
52.
Hirohata S, Miura Y, Tomita T, Yoshikawa H, Ochi T, Chiorazzi N. Enhanced
expression of mRNA for nuclear factor kappaB1 (p50) in CD34+ cells of the
bone marrow in rheumatoid arthritis. Arthritis Research & Therapy 2006;
8(2):R54.
53.
Marinova-Mutafchieva L, Williams RO, Funa K, Maini RN, Zvaifler NJ.
Inflammation is preceded by tumor necrosis factor-dependent infiltration of
mesenchymal cells in experimental arthritis. Arthritis and Rheumatism 2002;
46(2):507-513.
54.
van Eden W, Holoshitz J, Nevo Z, Frenkel A, Klajman A, Cohen IR. Arthritis
induced by a T-lymphocyte clone that responds to Mycobacterium tuberculosis
and to cartilage proteoglycans. Proceedings of the National Academy of
Sciences of the United States of America 1985; 82(15):5117-5120.
55.
Choe JY, Crain B, Wu SR, Corr M. Interleukin 1 receptor dependence of
serum transferred arthritis can be circumvented by toll-like receptor 4
signaling. The Journal of Experimental Medicine 2003; 197(4):537-542.
56.
Tanaka S. Regulation of bone destruction in rheumatoid arthritis through
RANKL-RANK pathways. World Journal of Orthopedics 2013; 4(1):1-6.
57.
Schett G. Cells of the synovium in rheumatoid athritis. Osteoclasts. Arthritis
Research & Therapy 2007; 9(1):203.
58.
Haugeberg G, Green MJ, Conaghan PG, Quinn M, Wakefield R, Proudman
SM, Stewart S, Hensor E, Emery P. Hand bone densitometry: a more sensitive
75
standard for the assessment of early bone damage in rheumatoid arthritis.
Annals of the Rheumatic Diseases 2007; 66(11):1513-1517.
59.
Stewart A, Mackenzie LM, Black AJ, Reid DM. Predicting erosive disease in
rheumatoid arthritis. A longitudinal study of changes in bone density using
digital X-ray radiogrammetry: a pilot study. Rheumatology (Oxford) 2004;
43(12):1561-1564.
60.
Gough AK, Peel NF, Eastell R, Holder RL, Lilley J, Emery P. Excretion of
pyridinium crosslinks correlates with disease activity and appendicular bone
loss in early rheumatoid arthritis. Annals of the Rheumatic Diseases 1994;
53(1):14-17.
61.
Als OS, Gotfredsen A, Riis BJ, Christiansen C. Are disease duration and
degree of functional impairment determinants of bone loss in rheumatoid
arthritis? Annals of the Rheumatic Diseases 1985; 44(6):406-411
62.
Lorenzo J, Choi Y, Horowitz M, Takayanagi H. Overview: The developing field
of osteoimmunology; in: Osteoimmunology: Interactions of the immune and
skeletal systems 2011; Chapter 1: Page 1-5. Published by Academic Press.
63.
Dénarié D, Constant E, Thomas T, Marotte H. Could biomarkers of bone,
cartilage or synovium turnover be used for relapse prediction in rheumatoid
arthritis patients? Mediators of Inflammation 2014; 2014: Article ID 537324.
64.
Christgau S, Garnero P, Fledelius C, Moniz C, Ensig M, Gineyts E, Rosenquist
C, Qvist P. Callagen type II C-telopeptide fragments as an index of cartilage
degradation. Bone 2001; 29(3):209-215.
65.
FitzGerald O, Soden M, Yanni G, Robinson R, Bresnihan B. Morphometric
analysis of blood vessels in synovial membranes obtained from clinically
affected and unaffected knee joints of patients with rheumatoid arthritis. Annals
of the Rheumatic Diseases 1991; 50(11):792-796.
66.
DeBusk LM, Chen Y, Nishishita T, Chen J, Thomas JW, Lin PC. Tie2 receptor
tyrosine kinase, a major mediator of tumor necrosis factor alpha-induced
angiogenesis in rheumatoid arthritis. Arthritis and Rheumatism 2003;
48(9):2461-2471.
76
67.
Marrelli A, Cipriani P, Liakouli V, Carubbi F, Perricone C, Perricone R,
Giacomelli R. Angiogenesis in rheumatoid arthritis: a disease specific process
or a common response to chronic inflammation? Autoimmunity Reviews 2011;
10(10):595-598.
68.
Halilova KI, Brown EE, Morgan SL, Bridges SL Jr, Hwang MH, Arnett DK,
Danila MI. Markers of treatment response to methotrexate in rheumatoid
arthritis: where do we stand? International Journal of Rheumatology 2012;
2012: Article ID 978396.
69.
Katchamart W, Johnson S, Lin HJL, Phumethum V, Salliot C, Bombardier C.
Predictors for remission in rheumatoid arthritis patients: a systematic review.
Arthritis Care and Research 2010; 62(8):1128-1143.
70.
Hider SL, Silman AJ, Thomson W, Lunt M, Bunn D, Symmons DP. Can clinical
factors at presentation be used to predict outcome of treatment with
methotrexate in patients with early inflammatory polyarthritis? Annals of the
Rheumatic Diseases 2009; 68(1):57-62.
71.
Van Vollenhoven RF, Ernestam S, Geborek P, Petersson IF, Cöster L,
Waltbrand E, Zickert A, Theander J, Thörner A, Hellström H, Teleman A,
Dackhammar C, Akre F, Forslind K, Liung L, Oding R, Chatzidionysiou A,
Wörnert M, Bratt J. Addition of infliximab compared with addition of
sulfasalazine and hydroxychloroquine to methotrexate in patients with early
rheumatoid arthritis (Swefot trial): 1-year results of a randomised trial. Lancet
2009; 374(9688):459-466.
72.
Saevarsdottir S, Wallin H, Seddighzadeh M, Ernestam S, Geborek P,
Petersson IF, Bratt J, van Vollenhoven RF; SWEFOT Trial Investigators
Group. Predictors of response to methotrexate in early DMARD naïve
rheumatoid arthritis: results from the initial open-label phase of the SWEFOT
trial. Annals of the Rheumatic Diseases 2011; 70(3):469-475.
73.
Mandl P, Balint PV, Brault Y, Backhaus M, D’Agostino MA, Grassi W, van der
Heijde D, de Miguel E, Wakefield Rj, Logeart I, Dougados M. Metrologic
properties of ultrasound versus clinical evaluation of synovitis in rheumatoid
arthritis: results of a multicenter, randomized study. Arthritis and Rheumatism
2012; 64(4):1272-1282.
77
74.
Taylor PC. Qualification of ultrasonography as a biomarker of prognosis and
response to treatment in early rheumatoid arthritis 2012. Available from
http://clinicaltrials.gov/ct2/show/NCT00781989?term=NCT00781989&rank=1
75.
Dohn UM, Ejbjerg B, Boonen A, Hetland ML, Hansen MS, Knudsen LS,
Hansen A, Madsen OR, Hasselquist M, Møller JM, Ostergaard M. No overall
progression and occasional
repair of erosions despite persistent
inflammation in adalimumab-treated rheumatoid arthritis patients: results from
a longitudinal comparative MRI, ultrasonography, CT and radiography study.
Annals of the Rheumatic Diseases 2011; 70(2):252-258.
76.
Goronzy JJ, Matteson EL, Fulbright JW, Warrington KJ, Chang-Miller A,
Hunder GG, Mason TG, Nelson AM, Valente RM, Crowson CS, Erlich HA,
Reynolds RL, Swee RG, O’Fallon WM, Weyand CM. Prognostic markers of
radiographic progression in early rheumatoid arthritis. Arthritis and
Rheumatism 2004; 50(1):43-54.
77.
Wagner U, Kaltenhäuser S, Sauer H, Arnold S, Seidel W, Häntzschel H,
Kalden JR, Wassmuth R. HLA markers and prediction of clinical course and
outcome in rheumatoid arthritis. Arthritis and Rheumatism 1997; 40(2):341351.
78.
Turesson C, Matteson EL. Genetics of rheumatoid arthritis. Mayo Clinic
Proceedings 2006; 81(1):94-101.
79.
Mori S, Hirose J, Yonemura K. Contribution of HLA-DRBI*04 alleles and anticyclic citrullinated antibodies to development of resistance to diseasemodifying antirheumatic drugs in early rheumatoid arthritis. Clinical
Rheumatology 2010; 29(12):1357-1366.
80.
O'Dell JR, Nepom BS, Haire C, Gersuk VH, Gaur L, Moore GF, Drymalski W,
Palmer W, Eckhoff PJ, Klassen LW, Wees S, Thiele G, Nepom GT. HLA-DRBI
typing in rheumatoid arthritis: predicting response to specific treatments.
Annals of the Rheumatic Diseases 1998; 57(4):209-213.
81.
Rodriguez-Rodriguez L, López-Mejias R, Garcia-Bermùdez M, GonzálezJuanatey C, González-Gay MA, Martin J. Genetic markers of cardiovascular
disease in rheumatoid arthritis. Mediators of Inflammation 2012; 2012: Article
ID 574817.
78
82.
González-Gay MA, González-Juanatey C, Lopez-Diaz MJ, Piñero A, GarciaPorrua C, Miranda-Filloy JA, Ollier WE, Martin J, Llorca J. HLA-DRBI and
persistent chronic inflammation contribute to cardiovascular events and
cardiovascular mortality in patients with rheumatoid arthritis. Arthritis and
Rheumatism 2007; 57(1):125-132.
83.
Farragher TM, Goodson NJ, Naseem H, Silman AJ, Thomson W, Symmons D,
Barton A. Association of the HLA-DRBI gene with premature death, particularly
from cardiovascular disease, in patients with rheumatoid arthritis and
inflammatory polyarthritis. Arthritis and Rheumatism 2008; 58(2):359-369.
84.
Mattey DL, Thomson W, Ollier WER, Batley M, Davies PG, Gough AK, Devlin
J, Prouse P, James DW, Williams PL, Dixey J, Winfield J, Cox NL, Koduri G,
Young A. Association of DRBI shared epitope genotypes with early mortality in
rheumatoid arthritis: results of eighteen years of follow up from the early
rheumatoid arthritis study. Arthritis and Rheumatism 2007; 56(5):1408-1416.
85.
Wesoly J, van der Helm-van Mil AH, Toes RE, Chokkalingam AP, Carlton VE,
Begovich AB, Huizinga TW. Association of the PTPN22 C1858T singlenucleotide polymorphism with rheumatoid arthritis phenotypes in an inception
cohort. Arthritis and Rheumatism 2005; 52(9):2948-2950.
86.
Plenge RM, Selelstad M, Padyukov L, Lee AT, Remmers EF, Ding B, Liew A,
KhaliliH, Chandrasekaran A, Davies LR, Li W, Tan AK, Bonnard C, Ong RT,
Thalamuthu A, Petersson S, Liu C, Tian C, Chen WV, Carulli JP,Beckman Em.
Altshuler D, Alfredsson L, Criswell LA, Amos CI, Seldin MF, Kastner DL,
Klareskog L, Gregersen PK. TRAFI-C5 as a risk locus for rheumatoid arthritis –
a genome wide study. The New England Journal of Medicine 2007;
357(10):1199-1209.
87.
Remmers EF, Plenge RM, Lee AT, Graham RR, Hom G, Behrens TW, de
Bakker PI, Le JM, Lee HS, Batliwalla F, Li W, Masters SL, Booty MG, Carulli
JP, Padyukov L, Alfredsson L, KlareskogL, Chen WV, Amos CI, Criswell LA,
Seldin MF, Kastner DL, Gregersen PK. STAT4 and the risk of rheumatoid
arthritis and systemic lupus erythematosus. The New England Journal of
Medicine 2007; 357(10):977-986.
79
88.
Chen Y, Nixon NB, Dawes PT, Mattey DL. Influence of variations across the
MMP-1 and -3 genes on the serum levels of MMP-1 and -3 and disease
activity in rheumatoid arthritis. Genes and Immunity 2012; 13(1):29-37.
89.
Sharma S, Das M, Kumar A, Marwaha V, Shankar S, Singh P, Raghu P, Aneja
R, Grover R, Arya V, Dhir V, Gupta R, Kumar U, Juyal RC, K TB. Purine
biosynthetic pathway genes and methotrexate response in rheumatoid arthritis
patients among north Indians. Pharmacogenetics and Genomics 2009;
19(10):823-828.
90.
Lee YC, Cui J, Costenbader KH, Shadick NA, Weinblatt ME, Karlson EW.
Investigation of candidate polymorphisms and disease activity in rheumatoid
arthritis patients on methotrexate. Rheumatology 2009; 48(6):613-617.
91.
Seitz M, Zwicker M, Villiger PM. Pretreatment cytokine profiles of peripheral
blood mononuclear cells and serum from patients with rheumatoid arthritis in
different american college of rheumatology response groups to methotrexate.
Journal of Rheumatology 2003; 30(1):28-35.
92.
Burska A, Boissinot M, Ponchel F. Cytokines as biomarkers in rheumatoid
arthritis. Mediators of Inflammation 2014; 2014, Article ID 545493.
93.
Burmester R. Does gene expression analysis inform us in rheumatoid arthritis?
Annals of the Rheumatic Diseases 2010; 69(Suppl 1):i37-i42.
94.
Lequerré T, Gauthier-Jauneau AC, Bansard C, Derambure C, Hiron M,
Vittecoq O,Daveau M, Mejjad O, Daragon A, Tron F, Le Loët X, Salier JP.
Gene profiling in white blood cells predicts infliximab responsiveness in
rheumatoid arthritis. Arthritis Research & Therapy 2006; 8(4):R105.
95.
Juliá A, Erra A, Palacio C, Tomas C, Sans X, Barceló P. Marsal S. An eightgene blood expression profile predicts the response to infliximab in rheumatoid
arthritis. PLoS One 2009; 4(10):e7556.
96.
Duroux-Richard I, Pers Y, Fabre S, Ammari M, Baeten D, Cartron G, Touitou I,
Jorgensen C, Apparailly F. Circulating miRNA-125b is a potential biomarker
predicting response to rituximab in rheumatoid arthritis. Mediators of
Inflammation 2014; 2014: Article ID 342524.
80
97.
Murata K, Yoshitomi H, Tanida S, IshikawaM, Nishitani K, Ito H, Nakamura T.
Plasma and synovial fluid microRNAs as potential biomarkers of rheumatoid
arthritis and osteoarthritis. Arthritis Research and Therapy 2010; 12(3):R86.
98.
Murata K, Furu M, Yoshitomi H, Ishikawa M,Shibuya H, Hashimoto M, Imura Y,
Fujii T, Ito H, Mimori T, Matsuda S. Comprehensive microRNA analysis
identifies miR-24 and miR-125a-5p as plasma biomarkers for rheumatoid
arthritis. PLoS One 2013; 8(7):e69118.
99.
Ingegnoli F, Castelli R, Gualtierotti R. Rheumatoid factors: clinical applications.
Disease Markers 2013; 35(6):727-734.
100. Nielsen SF, Bojesen SE, Schnohr P, Nordestgaard BG. Elevated rheumatoid
factor and long term risk of rheumatoid arthritis: a prospective cohort study.
British Medical Journal 2012; 345:e5244.
101. Barra L, Bykerk V, Pope JE, Haraoui BP, Hitchon CA, Thorne JC, Keystone
EC, Boire G; Catch Investigators. Anticitrullinated protein antibodies and
rheumatoid factor fluctuate in early inflammatory arthritis and do not predict
clinical outcomes. Journal of Rheumatology 2013; 40(8):1259-1267.
102. Hyrich KL, Watson KD, Silman AJ, Symmons DPM. Predictors of response to
anti-TNF-α therapy among patients with rheumatoid arthritis: results from the
British Society for Rheumatology Biologics Register. Rheumatology 2006;
45(12):1558-1565.
103. Bos WH, Bartelds GM, Wolbink GJ, de Koning MH, van de Stadt RJ, van
Schaardenburg D, Dijkmans BA, Nurmohamed MT. Differential response of
the rheumatoid factor and anticitrullinated protein antibodies during
adalimumab treatment in patients with rheumatoid arthritis. Journal of
Rheumatology 2008; 35(10):1972-1977.
104. Salgado E, Maneiro JR, Carmona L, Gomez-Reino J. Rheumatoid factor and
response to TNF antagonists in rheumatoid arthritis: systematic review and
meta-analysis of observational studies. Joint Bone Spine 2014; 81(1):41-50.
81
105. Klaasen R, Cantaert T, Wijbrandts CA, Teitsma C, Gerlag DM, Out TA, de
Nooijer MJ, Baeten D, Tak PP. The value of rheumatoid factor and anticitrullinated protein antibodies as predictors of response to infliximab in
rheumatoid arthritis: an exploratory study. Rheumatology 2011; 50(8):14871493.
106. Potter C, Hyrich KL, Tracey A, Lunt M, Plant D,Symmons DP, Thomson W,
Worthington J, Emery P, Morgan AW, Wilson AG, Isaacs J, Barton A;
BRAGGSS. Association of rheumatoid factor and anti-cyclic citrullinated
peptide positivity, but not carriage of shared epitope or PTPN22 susceptibility
variants, with anti-tumor necrosis factor response in rheumatoid arthritis.
Annals of the Rheumatic Diseases 2009; 68(1):69-74.
107. Bobbio-Pallavicini E, Caporali R, Alpini C, Moratti R, Montecucco C. Predictive
value of antibodies to citrullinated peptides and rheumatoid factors in anti-TNFα treated patients. Annals of the New York Academy of Sciences 2007;
1109:287-295.
108. Edwards JCW, Szczepański L, Szechiński J, Filipowicz-Sosnowska A,
EmeryP, Close DR, Stevens RM, Shaw T. Efficacy of B-cell-targeted therapy
with rituximab in patients with rheumatoid arthritis. The New England Journal
of Medicine 2004; 350(25):2572-2581.
109. Quartuccio L, Fabris M, Salvin S, Atzeni F, Saracco M, Benucci M, Cimmino
M, Morassi P, Masolini P, Pellerito R, Cutolo M, Puttini PS, De Vita S.
Rheumatoid factor positivity rather than anti-CCP positivity, a lower disability
and a lower number of anti-TNF agents failed are associated with response to
rituximab in rheumatoid arthritis. Rheumatology 2009; 48(12):1557-1559.
110. Garza-Garcia C, Rocio SS, Orea-Tejeda A, Castillo-Martínez L, Eduardo C,
López-Campos J, Keirns-Davis C. Risk factors for asymptomatic ventricular
dysfunction in rheumatoid arthritis patients. ISRN Cardiology 2013; 2013:
Article ID 635439.
111. Vizzardi E, Cavazzana I. Echocardiographic evaluation of asymptomatic
patients affected by rheumatoid arthritis. Journal of Investigative Medicine
2012; 60(8):1204-1208.
82
112. Kaplan MJ. Cardiovascular complications of rheumatoid arthritis: assessment,
prevention and treatment. Rheumatic Disease Clinics of North America 2010;
36(2):405-426.
113. Tomasson G, Aspelund T, Jonsson T, Valdimarsson H, Felson DT, Gudnason
V. The effect of rheumatoid factor on mortality and coronary heart disease.
Annals of the Rheumatic Diseases 2010; 69(9):1649-1654.
114. Aletaha D, Neogi T, Silman AJ, FunovitsJ, Felson DT, Bingham CO 3rd,
Birnbaum NS, Burmester GR, Bykerk VP, Cohen MD, Combe B, Costenbader
KH, Dougados M, Emery P, Ferraccioli G, Hazes JM, Hobbs K, Huizinga TW,
Kavanaugh A, Kay J, Kvien TK, Laing T, Mease P, Ménard HA, Moreland LW,
Naden RL, Pincus T, Smolen JS, Stanislawska-Biernat E, Symmons D, Tak
PP, Upchurch KS, Vencovský J, Wolfe F, Hawker G. 2010 Rheumatoid arthritis
classification criteria: an American College of Rheumatology/European League
Against Rheumatism collaborative initiative. Arthritis and Rheumatism 2010;
62(9):2569-2581.
115. Syversen SW, Gaarder PI, Goll GL, Ødegård S, Haavardsholm EA, Mowinckel
P, van derHeijde D, Landewé R, Kvien TK. High anti-cyclic citrullinate peptide
levels and an algorithm of four variables predict radiographic progression in
patients with rheumatoid arthritis: results from a 10-year longitudinal study.
Annals of the Rheumatic Diseases 2008; 67(2):212-217.
116. Van der Woude D. Syversen SW, van der Voort EIH, Verpoort KN, Goll GL,
van der Linden MP, van der Helm-van Mil AH, van der Heijde DM, Huizinga
TW, Kvien TK, Toes RE. The ACPA isotype
profile reflects long-term
radiographic progression in rheumatoid arthritis. Annals of the Rheumatic
Diseases 2010; 69(6):1110-1116.
117. Burska AN, Hunt L, Boissinot M, Strollo R, Ryan BJ, Vital E, Nissim A, Winyard
PG, Emery P, Ponchel F. Autoantibodies to post translational modifications in
rheumatoid arthritis. Mediators of Inflammation 2014; 2014, Article ID 492873.
118. Van der Helm-van Mil AHM, Verpoort KN, Breedveld FC, Toes REM, Huizinga
TWJ. Antibodies to citrullinated proteins and differences in clinical progression
of rheumatoid arthritis. Arthritis Research & Therapy 2005; 7(5):R949-R958.
83
119. Jansen LM, van Schaardenburg D, van der Horst-Bruinsma IE, van de Stadt
RJ, de Koning MHMT, Dijkmans BAC. The predictive value of anti-cyclic
citrullinated peptide antibodies in early arthritis. The Journal of Rheumatology
2003; 30(8):1691-1695.
120. Machold KP, Stamm TA, Nell VP, Pflugbeil S, Aletaha D, Steiner G,Uffmann
M, Smolen JS. Very recent onset rheumatoid arthritis: clinical and serological
patient characteristics associated with radiographic progression over the first
years of disease. Rheumatology 2007; 46(2):342-349.
121. Drynda S, Gloetzner M, Polter J, et al. Prediction of therapy response in antiTNFα treated RA patients based on the identification of biomarkers. Arthritis &
Rheumatology 2008; 58(Suppl 9):S211.
122. Isaacs JD, Olech E, Tak PP, Deodhar A, Keystone E, Emery P, Yocum D,
Hessey E, Read S. Autoantibody-positive rheumatoid arthritis (RA) patients
have enhanced clinical response to rituximab (RTX) when compared with
seronegative patients. Annals of the Rheumatic Diseases 2009; 68(Suppl
3):442.
123. Sellam J, Hendel-Chavez H, Rouanet S, Abbed K, Combe B, Le Loët X, Tebib
J, Sibilia J, Taoufik Y, Dougados M, Mariette X. B cell activation biomakers as
predictive factors for the response to rituximab in rheumatoid arthritis. Arthritis
& Rheumatology 2011; 63(4):933-938.
124. Vis M, Bos WH, Wolbink G, Voskuyl AE, Twisk JW, Van de Stadt R, Hamann
D, Dijkmans BA, Lems WF. IgM-rheumatoid factor, anti-cyclic citrullinated
peptide, and anti-citrullinated human fibrinogen antibodies decrease during
treatment with the tumor necrosis factor blocker infliximab in patients with
rheumatoid arthritis. The Journal of Rheumatology 2008; 35(3):425-428.
125. Cuchacovich M, Catalan D, Wainstein E, Gatica H, SotoL, Aravena O, Pesce
B, Sabugo F, Aguillón JC. Basal anti-cyclic citrullinated peptide (anti-CCP)
antibody levels and a decrease in anti-CCP titres are associated with clinical
response to adalimumab in rheumatoid arthritis. Clinical and Experimental
Rheumatology 2008; 26(6):1067-1073.
84
126. Atzeni F, Sarzi-Puttini P, Dell' Acqua D, de Portu S, Cecchini G, Cruini C,
Carrabba M, Meroni PL. Adalimumab clinical efficacy is associated with
rheumatoid factor and anticyclic citrullinated peptide antibody titer reduction: a
one-year prospective study. Arthritis Research & Therapy 2006; 8(1):R3.
127. Allanore Y, Sellam J, Batteux F, Job Deslandre C, Weill B, Kahan A. Induction
of autoantibodies in refractory rheumatoid arthritis treated by infliximab.
Clinical and Experimental Rheumatology 2004; 22(6):756-758.
128. Alessandri C, Priori R, Modesti M, Mancini R, Valesini G. The role of anti-cyclic
cytrullinate antibodies testing in rheumatoid arthritis. Clinical Reviews in
Allergy & Immunology 2008; 34(1):45-49.
129. Chen HA, Lin KC, Chen CH, Liao HT, Wang HP, Chang HN, Tsai CY, Chou
CT. The effect of etanercept on anti-cyclic citrullinated peptide antibodies and
rheumatoid factor in patients with rheumatoid arthritis. Annals of the
Rheumatic Diseases 2006; 65(1):35-39.
130. Alessandri C, Bombardieri M, Papa N, Cinguini M, Magrini L, Tincani A,
Valesini G. Decrease of anti-cyclic citrullinated peptide antibodies and
rheumatoid factor following anti-TNFα therapy (infliximab) in rheumatoid
arthritis is associated with clinical improvement. Annals of the Rheumatic
Diseases 2004; 63(10):1218-1221.
131. Benucci M, Turchini S, Parrochi P, Boccaccini P, Manetti R, Cammelli E,
ManfrediM. [Correlations between different clinical activity and anti CC-P (anticyclic citrullinated peptide antibodies) titres in rheumatoid arthritis treated with
three different tumor necrosis factors TNF-α blockers]. Recenti Progressi in
Medicina 2006; 97(3):134-139. [Article in Italian].
132. Braun-Moscovici Y, Markovits D, Zinder O, Schapira D, Rozin A, Ehrenburg M,
Dain L, Hoffer E, Nahir AM, Balbir-Gurman A. Anti-cyclic citrullinated protein
antibodies as a predictor of response to anti-tumor necrosis factor-α therapy in
patients with rheumatoid arthritis. The Journal of Rheumatology 2006;
33(3):497-500.
133. Bobbio-Pallavicini F, Alpini C, Caporali R, Avalle S, Bugatti S, Montecucco C.
Autoantibody profile in rheumatoid arthritis during long-term infliximab
treatment. Arthritis Research & Therapy 2004; 6(3):R264-R272.
85
134. Taylor P, Gartemann J, Hsieh J, Creeden J. A systematic review of serum
biomarkers anti-cyclic citrullinated peptide and rheumatoid factor as tests for
rheumatoid arthritis. Autoimmune Diseases 2011; 2011: Article ID 815038.
135. Bizzaro N. Antibodies to citrullinated peptides: a significant step forward in the
early diagnosis of rheumatoid arthritis. Clinical Chemistry and Laboratory
Medicine 2007; 45(2):150-157.
136. De Rycke L, Verhelst X, Kruithof E, Van den Bosch F, Hoffman IE, Veys EM,
De Keyser F. Rheumatoid factor, but not anti-cyclic citrullinated peptide
antibodies, is modulated by infliximab treatment in rheumatoid arthritis. Annals
of the Rheumatic Diseases 2005; 64(2):299-302.
137. Teng Y, Verburg RJ, Verpoort KN, Diepenhorst GM, Bajema IM, van Tol MJ,
Jol-van der Zijde EC, Toes RE, Huizinga TW, van Laar JM. Differential
responsiveness to immunoablative therapy in refractory rheumatoid arthritis is
associated with level and avidity of anti-cyclic citrullinated protein
autoantibodies: a case study. Arthritis Research & Therapy 2007; 9(5):R106.
138. Gerli R, Bartoloni Bocci E, Sherer Y, Vaudo G, Moscatelli S, Shoenfeld Y.
Association of anti-cyclic citrullinated peptide antibodies with subclinical
atherosclerosis in patients with rheumatoid arthritis. Annals of the Rheumatic
Diseases 2008; 67(5):724-725.
139. Lopez-Longo FJ, Oliver-Minarro D, de la Torre I, González-Diaz de Rábago E,
Sánchez-Ramón S, Rodríguez-Mahou M, Paravisini A, Monteagudo I,
González CM, García-Castro M, Casas MD, Carreño L. Association between
anti-cyclic citryllinated peptide antibodies and ischemic heart disease in
patients with rheumatoid arthritis. Arthritis & Rheumatology 2009; 61(4):419424.
140. Trouw LA, Huizinga WJ, Toes REM. Autoimmunity in rheumatoid arthritis:
different antigens - common principles. Annals of the Rheumatic Diseases
2013; 72(Suppl 2):ii132-ii136.
141. Wang Z, Nicholis SJ, Rodriguez ER, Kummu O, Hörkkö S, Barnard J,
Reynolds WF, Topol EJ, DiDonato JA, Hazen SL.. Protein carbamylation links
inflammation, smoking, uremia and atherogenesis. Nature Medicine 2007;
13(10):1176-84.
86
142. Sirpal S. Myeloperoxidase-mediated lipoprotein carbamylation as a
mechanistic pathway for atherosclerotic vascular disease. Clinical Science
2009; 116(9):681-695.
143. Fardellone P, Séjourné A, Paccou J, Goëb V. Bone remodelling markers in
rheumatoid arthritis. Mediators of Inflammation 2014; 2014, Article ID 484280.
144. Forsblad d'Elia H, Larsen A, Waltbrand E, Kvist G, Mellström D, Saxne T,
Ohlsson C, Nordborg E, Carlsten H. Radiographic joint destruction in
postmenopausal rheumatoid arthritis is strongly associated with generalised
osteoporosis. Annals of the Rheumatic Diseases 2003; 62(7):617-623.
145. Jensen T, Hansen M, Jensen KE, Podenphant J, Hansen TM, Hyldstrup L.
Comparison of dual X-ray absorptiometry (DXA), digital X-ray radiogrammetry
(DXR), and conventional radiographs in the evaluation of osteoporosis and
bone erosions in patients with rheumatoid arthritis. Scandinavian Journal of
Rheumatology 2005; 34(1):27-33.
146. Szentpetery A, McKenna MJ, Murray BF, Nq CT, Brady JJ, Morrin M, Radovits
B, Veale DJ, Fitzgerald O. Periarticular bone gain atproximal interphalangeal
joints and changes in bone turnover makers in response to tumor necrosis
factor inhibitors in rheumatoid and psoriatic arthritis. The Journal of
Rheumatology 2013; 40(5):653-662.
147. Terpos E, Fragiadaki K, Konsta M, Brtengeier C, Papatheodorou A, Sfikakis
PP. Early effects of IL-6 receptor inhibition on bone homeostasis: a pilot study
in women with rheumatoid arthritis. Clinical and Experimental Rheumatology
2011; 29(6):921-925.
148. Van Ruyl LHD, Voskuyl AE, Boers M, Geusens P, Landewé RB, Dijkmans BA,
Lems WF. Baseline RANKL: OPG ratio and markers of bone and cartilage
degradation predict annual radiological progression over 11 years in
rheumatoid arthritis. Annals of the Rheumatic Diseases 2010; 69(9):16231628.
149. Kanbe K, Nakamura A, Inoue Y, Hobo K. Osteoprotegerin expression in bone
marrow by treatment with tocilizumab in rheumatoid arthritis. Rheumatology
International 2012; 32(9):2669-2674.
87
150. Vis M, Havaardsholm EA, Haugeberg G, Uhlig T, Voskuyl AE, van de Stadt
RJ, Dijkmans BA, Woolf AD, Kvien TK, Lems WF. Evaluation of bone mineral
density, bone metabolism, osteoprotegerin and receptor activator of the NFkB
ligand serum levels during treatment with infliximab in patients with rheumatoid
arthritis. Annals of the Rheumatic Diseases 2006; 65(11):1495-1499.
151. Yamanaka H, Matsuda Y, Tanaka M, Sendo W, Nakajima H, Taniguchi A,
Kamatani N. Serum matrix metalloproteinase 3 as a predictor of the degree of
joint destruction during the six months after measurement, in patients with
early rheumatoid arthritis. Arthritis & Rheumatology 2000; 43(4):852-858.
152. Funahashi K, Koyano S, Miura T, Hagiwara T, Okuda K, Matsubara T. Efficacy
of tocilizumab and evaluation of clinical remission as determined by CDAI and
MMP-3 level. Modern Rheumatology 2009; 19(5):507-512.
153. Nishimoto N, Amano K, Hirabayashi Y, horiuchi T, Ishii T, Iwahashi M,
Iwamoto M, Kohsaka H, Kondo M, Matsubara T, Mimura T, Miyahara H, Ohta
S, Saeki Y, Saito K, Sano H, Takasugi K, Takeuchi T, Tohma S, Tsuru T, Ueki
Y, Yamana J, Hashimoto J, Matsutani T, Murakami M, Takagi N. Drug free
REmission/low disease activity after cessation of tocilizumab (Actemra)
Monotherapy (DREAM) study. Modern Rheumatology 2014; 24(1):17-25.
154. Houseman M, Potter C, Marshall N, Lakey R, Cawston T, Griffiths I, YoungMin S, Isaacs JD. Baseline serum MMP-3 levels in patients with rheumatoid
arthritis are still independently predictive of radiographic progression in a
longitudinal observational cohort at 8 years follow up. Arthritis Research and
Therapy 2012; 14(1):R30.
155. Urata Y, Uesato R, Tanaka D, Nakamura Y Motomura S. Treating to target
matrix metalloproteinase 3 normalisation together with disease activity score
below 2.6 yields better effects than each alone in rheumatoid arthritis patients:
T-4 Study. Annals of the Rheumatic Diseases 2012; 71(4):534-540.
156. Visvanathan S, Marini JC, Smolen JS, Clair EW, Pritchard C, Shergy W,
Pendley C, Baker D, Bala M, Gathany T, Han J, Wagner C. Changes in
biomarkers of inflammation and bone turnover and associations with clinical
efficacy following infliximab plus methotrexate therapy in patients with early
rheumatoid. The Journal of Rheumatology 2007; 34(7):1465-1474.
88
157. Morozzi G, Fabbroni M, Bellisai F, Cucini S, Simpatico A, Galeazzi M. Low
serum level of COMP, a cartilage turnover marker, predicts rapid and high
ACR70 response to adalimumab therapy in rheumatoid arthritis. Clinical
Rheumatology 2007; 26(8):1335-1338.
158. Johansen JS, Stoltenberg M, Hansen M, Florescu A, Hørsley-Petersen K,
Lorenzen I, Price PA. Serum YKL-40 concentrations in patients with
rheumatoid arthritis: relation to disease activity. Rheumatology. 1999;
38(7):618-626.
159. Johansen JS, Jensen HS, Price PA. A new biochemical marker for joint injury.
Analysis of YKL-40 in serum and synovial fluid. British Journal of
Rheumatology 1993; 32(11):949-955.
160. Knudsen LS, Klarlund M, Skjɸdt H, Jensen T, Ostergaard M, Jensen KE,
Hansen MS, HetlandML, Nielsen HJ, Johansen JS. Biomarkers of
inflammation in patients with unclassified polyarthritis and early rheumatoid
arthritis. Relationship to disease activity and radiographic outcome. The
Journal of Rheumatology 2008; 35(7):1277-1287.
161. Landewé R, Geusens P, Boers M, et al. Markers for type II collagen
breakdown predict the effect of disease-modifying treatment on long-term
radiographic progression in patients with rheumatoid arthritis. Arthritis &
Rheumatology 2004; 50(5):1390-1399.
162. Straburzyńska-Lupa A, Nowak A, Romanowski W, Korman P, PilaczyńskaSzcześniak L. A study of the link between bone turnover markers and bone
mineral density with inflammation and body mass in postmenopausal women
with active rheumatoid arthritis. Journal of Bone and Mineral Metabolism 2013;
31(2):169-176.
163. Carnero P, Landewé R, Boers M, et al. Association of baseline levels of
markers of bone and cartilage degradation with long-term progression of joint
damage in patients with early rheumatoid arthritis: the COBRA study. Arthritis
& Rheumatology 2002; 46(11):2847-2856.
164. Karsdal MA, Schett G, Emery P, et al. IL-6 receptor inhibition positively
modulates bone balance in rheumatoid arthritis patients with an inadequate
89
response to anti-tumor necrosis factor therapy: biochemical marker analysis of
bone metabolism in the tocilizumab RADIATE study (NCT00106522).
Seminars in Arthritis and Rheumatism 2012; 42(2):131-139.
165. Hueber W, Tomooka BH, Zhao X, et al. Proteomic analysis of secreted
proteins in early rheumatoid arthritis: anti-citrulline autoreactivity is associated
with up regulation of proinflammatory cytokines. Annals of the Rheumatic
Diseases 2007; 66(6):712-719.
166. Hitchon CA, Alex P, Erdile LB, et al. A distinct multicytokine profile is
associated with anti-cyclical citrullinated peptide antibodies in patients with
early untreated inflammatory arthritis. The Journal of Rheumatology 2004;
31(12):2336-2346.
167. Shimamoto K, Ito T, Ozaki Y, et al. Serum interleukin 6 before and after
therapy with tocilizumab is a principal biomarker in patients with rheumatoid
arthritis. The Journal of Rheumatology 2013; 40(7):1074-1081.
168. Joosten LAB, Radstake TRD, Lubberts E, et al. Association of interleukin-1β
and tumor necrosis factor α in knee synovial tissue of patients with rheumatoid
arthritis. Arthritis & Rheumatology 2003; 48(2):339-347.
169. Van Roon JA, Jacobs K, Verstappen S, Bijlsma J, Lafeber F. Reduction of
serum interleukin 7 levels upon methotrexate therapy in early rheumatoid
arthritis correlates with disease suppression. Annals of the Rheumatic
Diseases 2008; 67(7):1054-1055.
170. Macías I, García-Pérez S, Ruiz-Tudela M, Medina F, Chozas N, GirónGonzález JA. Modification of pro- and anti-inflammatory cytokines and
vascular-related molecules by tumor necrosis factor-a blockade in patients with
rheumatoid arthritis. The Journal of Rheumatology 2005; 32(11):2102-2108.
171. Nishina N, Kaneko Y, Kameda H, Kuwana M, Takeuchi T. Reduction of
plasma IL-6 but not TNF-α by methotrexate in patients with early rheumatoid
arthritis: a potential biomarker for radiographic progression. Clinical
Rheumatology 2013; 32(11):1661-1666.
90
172. Goëb V, Aegerter P, Parmar R, Fardellone P, Vittecog O, Conaghan PG,
Emery P,Le Loët X, Ponchel F. Progression to rheumatoid arthritis in early
inflammatory arthritis is associated with low IL-7 serum levels. Annals of the
Rheumatic Diseases 2013; 72(6):1032-1036.
173. Mailefert JF, Puéchal X, Falgarone G, Lizard G, Ornetti P, Solau E, Legré V,
Lioté F, Sibilia J, Morel J, Maynadié M; Réseau Rhumato Study Group.
Prediction of response to disease modifying antirheumatic drugs in rheumatoid
arthritis. Joint Bone Spine 2010; 77(6):558-563.
174. Kasama T, Isojima S, Umemura M, Tsukamoto H, Tokunaga T, Furuya H,
Yanai R, Takahashi R, Nakamura M, Inagaki K. Serum macrophage migration
inhibitory factor levels are correlated with response to tocilizumab therapy in
patients with rheumatoid arthritis. Rheumatology International 2013; 34(3):429433.
175. Fabre S, Dupuy AM, Dossat N, Guisset C, Cohen JD, Cristol JP, Daures JP,
Jorgensen C. Protein biochip array technology for cytokine profiling predicts
etanercept responsiveness in rheumatoid arthritis. Clinical & Experimental
Immunology 2008; 153(2):188-195.
176. Fabre S, Guisset C, Tatem L, Dossat N, Dupuy AM, Cohen JD, Cristol JP,
Daures JP, Jorgensen C. Protein biochip array technology to monitor rituximab
in rheumatoid arthritis. Clinical & Experimental Immunology 2009; 155(3):395402.
177. Zivojinovic SM, Pejnovic NN, Sefik-Bukilica MN, Kovacevic LV, Soldatovic II,
Damjanov NS. Tumor necrosis factor blockade differentially affects innate
inflammatory and Th17 cytokines in rheumatoid arthritis. The Journal of
Rheumatology 2012; 39(1):18-21.
178. Hollander AP, Corke KP, Freemont AJ, Lewis CE. Expression of hypoxiainducible factor 1alpha by macrophages in the rheumatoid synovium:
implications for targeting of therapeutic genes to the inflamed joint. Arthritis
and Rheumatism 2001; 44(7):1540-1544.
91
179. Lee SS, Joo YS, Kim WU, Min DJ, Min JK, Park SH, Cho CS, Kim HY.
Vascular endothelial growth factor levels in the serum and synovial fluid of
patients with rheumatoid arthritis. Clinical and Experimental Rheumatology
2001; 19(3):321-324.
180. Paleolog EM, Young S, Stark AC, McCloskey RV, Feldmann M, Maini RN.
Modulation of angiogenic vascular endothelial growth factor by tumor necrosis
factor alpha and interleukin-1 in rheumatoid arthritis. Arthritis and Rheumatism
1998; 41(7):1258-65.
181. Dessein PH, Joffe BI, Singh S. Biomarkers of endothelial dysfunction,
cardiovascular risk factors and atherosclerosis in rheumatoid arthritis. Arthritis
Research & Therapy 2005; 7(3):R634-643.
182. Khan F, Galarraga B, Belch JJ. The role of endothelial function and its
assessment in rheumatoid arthritis. Nature Reviews Rheumatology 2010;
6(5):253-261.
183. Kilic T, Ural D, Ural E, yumuk Z, Agacdiken A, Sahin T, Kahraman G, Kozdag
G, Vural A, Komsuoglu B. Relation between proinflammatory to antiinflammatory cytokine ratios and long-term prognosis in patients with non-ST
elevation acute coronary syndrome. Heart 2006; 92(8):1041-1046.
184. Roman MJ, Devereux RB, Schwartz JE, Lockshin MD, Paget SA, Davis A,
Crow MK, Sammaritano L, Levine DM, Shankar Ba, Moeller E, Salmon JE.
Arterial stiffness in chronic inflammatory diseases. Hypertension 2005;
46(1):194-199.
185. Rho YH, Chung CP, Oeser A, Solus J, Asanuma Y, Sokka T, Pincus T, Raggi
P, Gebretsadik T, Shintani A, Stein CM. Inflammatory mediators and
premature coronary atherosclerosis in rheumatoid arthritis. Arthritis &
Rheumatology 2009; 61(11):1580-1585.
186. Shen Y, Cavet G, Dirven L, CentolaM, Dijkmans B, HesterbergL,Kerstens P,
Huizinga T, Allaart CF. Serum biomarkers predict progressive structural
damage in the BeSt study. Annals of the Rheumatic Diseases 2010; 69(Suppl
3):501.
92
187. Curtis JR, Haney D, van der Helm AHM, Shen Y, Knevel R, Cavet G, Dirven L.
Validation of a multi-biomarker test for rheumatoid arthritis (RA) disease
activity (Vectra DA) in a multi-cohort study. [abstract]. Arthritis & Rheumatology
2010; 62(Suppl 10):1782.
188. Hambardzumyan K, Bolce R, Saevarsdottir S, Cruickshank SE, Sasso EH,
Chernoff D, ForslindK, Petersson IF, Geborek P, van Vollenhoven RF.
Pretreatment multi-biomarker disease activity score and radiographic
progression in early RA: results from the SWEFOT trial. Ann Rheum Dis. 2014
May 8. [Epub ahead of print] [doi: 10.1136/annrheumdis-2013-204986]
189. Chopra A, Abdel-Nasser A. Epidemiology of rheumatic musculoskeletal
disorders in the developing world. Best Practice & Research Clinical
Rheumatology 2008; 22(4):583-604.
190. Symmons D, Turner G, Webb R, Asten P, Barrett E, Lunt M, Scott D, SilmanA.
The prevalence of rheumatoid arthritis in the United Kingdom: new estimates
for a new century. Rheumatology (Oxford) 2002; 41(7):793–800.
191. Helmick CG, Felson DT, Lawrence RC, Gabriel S, Hirsch R, Kwoh CK, Liang
MH, Kremers HM, Mayes MD, Merkel PA Pillemer SR, Reveille JD, Stone JH;
National Arthritis Data Workgroup. Estimates of the prevalence of arthritis
and other rheumatic conditions in the United States. Part I. Arthritis and
Rheumatism 2008; 58(1): 15–25.
192. Modi GM,Cardiel MH. Challenges in the management of rheumatoid arthritis in
developing countries: Best Practice & Research Clinical Rheumatology 2008;
22(4):621-641.
193. Adebajo AO, Reid DM. The pattern of rheumatoid arthritis in West Africa and
comparison with a cohort of British patients. The Quarterly Journal of Medicine
1991; 80(292):633–640.
194. Tikly M, Zannettou N, Hopley M. A longitudinal study of rheumatoid arthritis in
South Africans. Medscape General Medicine 2003; 5(1):2.
93
195. Hodkinson B, Musenge E, Ally M, Meyer PW, Anderson R, Tikly M. Functional
disability and health-related quality of life in South Africans with early
rheumatoid arthritis. Scandinavian Journal of Rheumatology 2012; 41(5):366374.
196. Okoye RC, Ollier W, Jaraquemada D, Awad J, Navarrete C, Cutbush S, Carthy
D, Dos-Santos A, Festenstein H. HLA-D region heterogeneity in a Nigerian
population. Tissue Antigens 1989; 33(4):445–456.
197. Pile KD, Tikly M, Bell JI, Wordsworth BP. HLA-DR antigens and rheumatoid
arthritis in black South Africans: a study of ethnic groups. Tissue Antigens
1992; 39(3):138–140.
198. Massardo L, Aguirre V, Garcia ME, Cervila V, Nicovani S, González A, Rivero
S, Jacobelli S. Clinical expression of rheumatoid arthritis in Chilean patients.
Seminars in Arthritis and Rheumatism 1995; 25(3):203–213.
94
CHAPTER 2
Patients and Methods
2.1
Study design
A prospective observational study conducted over two years recruited 171 patients
from the rheumatology clinics of two public sector tertiary academic hospitals, Chris
Hani Baragwanath and Steve Biko Academic Hospitals. Informed written consent was
obtained from all participants before recruitment into the study. Patients recruited met
the following inclusion and exclusion criteria:
Inclusion criteria
-
Fulfillment of the 1987 revised American College of Rheumatology
classification criteria for rheumatoid arthritis (at least 4 out of 7 criteria Table 1).
-
Age >18 years of age.
-
<2 years duration of disease (onset of characteristic articular symptoms
to enrolment less than 2 years)
-
Informed consent.
-
Disease-modifying anti-rheumatic drug (DMARD) therapy-naïve.
Exclusion criteria
-
Corticosteroid drugs in the preceding month (4 weeks) – intramuscular or
intravenous (stable oral dose for at least 2 weeks prior to enrollment at a dose
of ≤ 10mg daily was permitted).
-
Presence of co-existing overlap with any other causes of inflammatory arthritis
such as gout, connective tissue disorders, spondyloarthropathy or reactive
arthritis.
-
Chronic pain syndromes such as fibromyalgia.
95
Table 1: 1987 American College of Rheumatology (ACR) classification criteria for
rheumatoid arthritis.1
Classification Criteria
2.2
1
Morning stiffness for ≥1 hour
2
Arthritis of ≥3 joint areas
3
Arthritis of hand joints
4
Symmetric arthritis
5
Rheumatoid nodules
6
Serum rheumatoid factor (IgM)
7
Radiographic changes
Clinical and radiographic methods
At baseline, demographic data, duration of symptoms, current smoking history,
education level, medications including alternative therapies, laboratory tests, X-rays
of hands/feet and disease activity scores were recorded (joint counts were done by
experienced doctors from the rheumatology clinics). Clinical recording included extraarticular manifestations and noting the presence of any infections. Clinical parameters
were assessed at 6 monthly intervals including documentation of all therapeutic
changes (Appendix 2)
96
2.2.1
Disease activity variables assessed
-
Duration of early morning stiffness.
-
Tender joint count
-
Swollen joint count
-
Patient pain assessment (Likert scale).
-
Patient global assessment (Likert scale)
-
Physician global assessment (Likert scale)
-
Composite Disease Activity score (DAS 28)
-
Simplified Disease Activity Index (SDAI)
2.2.2 Functional assessment included
-
Modified Health Assessment Questionnaire (HAQ)2
-
Short Form-36 (SF-36) – Medical Outcome Study SF 363
2.2.3 Radiology
-
Radiographs were scored independently by two qualified rheumatologists in
chronological sequence applying the Larsen scoring method after achieving
acceptable inter-observer correlation. X-rays were also evaluated for the
presence of erosive disease and were performed at baseline and at yearly
intervals.
2.3
Serum
Venous blood (30ml) was collected in endotoxin-free, silicone-coated vacutainers
containing a gel separator. The blood samples were allowed to stand at room
temperature (±15 minutes) followed by centrifugation (3000 rpm for 10 minutes) after
which the serum was removed, aliquoted and stored at
-20oC until performance of
the various assays described below.
97
2.3.1 Rheumatoid Factor (RF)
RF (composite IgM, IgG, IgA), C-reactive protein (CRP) and serum amyloid A (SAA)
were assayed by immuno-nephelometry (Siemens Healthcare Diagnostics, BN
Prospec Nephelometer, Newark, USA) using reagents, standards and controls from
Siemens Healthcare Diagnostics. Immunonephelometry is the determination of the
concentration of a protein, by interaction of the protein with specific antisera to form
antibody-antigen complexes. The amount of light-scatter caused when a beam of light
(usually a red laser diode is the light source) is directed through the immune
complexes is measured by a detector and compared to the light scatter intensities of
a standard curve with known concentrations of the analyte. Serum results for RF,
CRP and SAA were considered positive when the concentrations were greater than
11 IU/ml, 5µg/ml and 6.8 µg/ml respectively.
2.3.2 Anti-citrullinated peptide antibodies (ACPA)
ACPA were measured by fluorescence enzyme immune assay (FEIA). FEIA utilizes
an antigen cyclic citrullinated peptide 2 (CCP2) bound to a solid phase (Immunocaps)
to capture the anti-CCP2 in serum. The bound antibody is then detected and
quantified using a fluorochrome-conjugated anti-human IgG according to the
magnitude of the florescence intensity using the Immunocap 250 system and
reagents, standards and controls from the manufacturer (Phadia AB, Uppsala,
Sweden). A result was considered positive for ACPA antibodies when the
concentration was >10 U/ml.
2.3.3 Antibodies to modified citrullinated vimentin (anti-MCV)
Anti-MCV levels were measured by enzyme-linked immunosorbent assay (ELISA).
Modified citrullinated vimentin (MCV) bound to a polypropylene matrix (flat-bottom 8well strips), was used to capture and measure anti-MCV present in serum following
the binding of enzyme-conjugated anti-human IgG to the anti-MCV in the presence of
a colorimetric substrate (Orgentec Diagnostika GmbH, Mainz, Germany). A serum
result was considered positive for anti-MCV antibodies when the concentration was
>20 U/ml.
98
2.3.4 Genotyping
Genomic DNA was obtained using the Promega Maxwell Personal Automation
System with the Maxwell 16 Blood DNA Purification Kit (Promega Corporation,
Madison, Wisconsin, USA) for extracting DNA from whole blood, collected in ethylene
diamine tetra acetic acid (EDTA) sample tubes. The yield and purity of DNA extracted
from EDTA blood samples were determined spectrophotometrically at 260/320nm
and 260/280nm respectively and the concentration of DNA adjusted to 50ng/µl as
required.
Genotyping of HLA-DRB1 alleles was performed using a DNA-based high-resolution
typing method, LABTypeHD DRB1 (One Lambda Inc, Canoga Park, California,
USA), utilising reverse sequence specific oligonucleotide (rSSO) Luminex xMAP
technology probes. Firstly, the target DNA (HLA-DRB1 gene) is amplified by PCR
using group- specific primers that are biotinylated for detection with SAPE (Rphycoerythrin-conjugated streptavidin).
The polymerase chain reaction (PCR)
product is then denatured and hybridised to complementary DNA probes conjugated
to fluorophores. Bound DNA is detected on the Luminex system by addition of SAPE,
and software maps the reaction patterns to those associated with published HLA
gene sequences, and assigns the represented HLA-DRB1 alleles.
Firstly, HLA
alleles were assigned according to the RAA amino-acid sequence at positions 72-74
as shared epitope (S) and non-SE (X), and the S group further subdivided according
to amino-acid residues at positions 70 and 71 as described by du Montcel4 (see
Table 2), with the groups pooled according to SE status into risk-allele-positive
(S2,S3P) and -negative (S1,S3D,X).5
99
Table 2: Shared Epitope classification according to amino acid sequences at
positions 70-74 4
Allele
Amino acid
HLA-DRB1 alleles
Classification sequence
position 70-74
S1
D-E-RAA
*01:03, *04:02, *11:02-03, *13:01-:02, *13:04,
*13:36, *13:40
S2
S3D
Q-A-RAA
*15:xx
Q-K-RAA
*04:01, 04:09, *04:13, *04:35, *04:66
D-K-RAA
*13:03
D-R-RAA
*11:01,
*11:04,
11:27,
*12:xx,
*13:05-:06,
*13:25, *14:22, *16
S3P
X
Q-R-RAA
*01:01-:02, *04:04-:05, *04:08, *04:10
R-R-RAA
*10:01
Q-K-RGR
*03:xx
Q-R-RAE
*04:03, *04:07, *04:11
D-R-RGQ
*07:xx
D-R-RAL
*08:xx
R-R-RAE
*09:01, *14:01, *14:04
2.3.5 Cytokines
Cytokines were measured using the Bio-Plex® suspension array system (Bio-Rad
 m ultiple x
Laboratories Inc, Hercules, CA, USA) which utilises Luminex® xMAP
technology to enable simultaneous detection and quantitation of multiple different
analytes in a single sample. The system uses an array of microspheres in liquid
suspension, conjugated with a monoclonal antibody specific for a target protein. The
beads contain different ratios of two spectrally distinct fluorophores, thereby assigning
a unique spectral identity. These antibody-coupled, colour-coded beads were then
100
incubated with the serum sample (¼ dilutions), washed, followed by addition of a
biotinylated detection antibody, washed again, and finally incubated with streptavidinphycoerythrin. A wide range of standards (0.38 – 91756.00 pg/ml) was used to
enable quantitation of the individual cytokines using a Bio-Plex array reader with a
dual laser detector and real time digital signal processing, in a fluidic system that
detects a single sphere. The following analytes were measured: IL-1, IL-1Ra, IL-2, IL4, IL-6, IL-7, IL-8, IL-10, IL-12, IL-17, IFN-γ, TNF, G-CSF, GM-CSF, CCL2, CCL4,
and VEGF. Ten healthy control subjects (5 female, 5 male, average age 44.4±15.0
years, ranging from 26 -65 years of age) were used to determine the upper normal
limit of the cytokine concentrations by calculating the mean +1SD.
2.3.6 Serum matrix metalloproteinase-3 (MMP-3) and cartilage oligomeric matrix
protein (COMP)
Serum concentrations of MMP-3 were measured using the Quantikine® Total MMP-3
Immunoassay (ELISA) according to the instructions supplied by the manufacturer (R
& D Systems, Minneapolis, MN, USA) and the results expressed as nanograms
(ng)/ml serum. Serum levels were measured in healthy controls (15 females and 10
males, age range 24-64). COMP was measured using the Kamiya Human COMP
ELISA system (Kamiya Biomedical Company, Seattle, WA, USA) and the results
expressed as µg/ml serum. Serum levels were also measured in 8 adult control
subjects.
2.4
Treatment
Patients were treated as per routine standard of care in the rheumatology units and
evaluated clinically at 2-4 monthly intervals. Pharmacological management included
monotherapy or combination therapy with methotrexate (MTX), salazopyrin (SZP)
and/or chloroquine (CQ) with or without corticosteroids. Choice of pharmaco-therapy
and dosage adjustments were at the discretion of the attending clinician as per
routine care. Methotrexate doses were initiated at 7.5 to 15 mg weekly and titrated up
to a maximum of 25 mg weekly. SZP was started at 500 mg daily and increased to 1g
twice a day with CQ prescribed at 200 mg daily.
101
2.5
Statistical Analysis
Descriptive statistics of continuous variables were performed by using a measure of
central tendency (mean or median) and a measure of variability (standard deviation
or range). Frequencies and percentages were used to describe categorical variables
of the nominal or ordinal scales. Cytokine data were not normally distributed and
therefore the Spearman’s pairwise correlations were used to find the strengths of
association of each cytokine with demographics and other biomarkers, as well as
between themselves. Medians and ranges were used to describe the skewed
numerical clinical and biomarker data. Non-parametric procedures were used in the
bivariate analysis of these data. The Mann-Whitney test was used to assess
differences in medians for two independent groups, while the Wilcoxon signed-rank
test was used to compare two dependent groups (i.e. baseline vs six months of
therapy). The Kruskal-Wallis test was used to determine differences in medians of
more than three independent groups. Statistical significance was ascertained at the
5% level i.e a p-value of less than 0.05 was deemed significant, and, where
appropriate, Bonferroni corrections to the p-values for multiple tests were applied.
Statistical analysis was done using STATA version 12.0 (Stata Corporation, College
Station, Texas USA).
2.6
Ethics
The study was approved by the Research Ethics Committees of the Faculties of
Health Sciences of the University of Pretoria and University of the Witwatersrand.
(protocol number 173/2010)
102
2.7
Funding obtained from
-
Connective Tissue Research Fund, University of the Witwatersrand.
-
Rheumatology Research Fund, Chris Hani Baragwanath Hospital.
-
Department of Immunology, University of Pretoria and National Health
Laboratory Service.
-
Rheumatology Research Fund, University of Pretoria and Steve Biko
Academic Hospital.
-
Medical Research Council of South Africa.
103
References
1. Arnett FC, Edworthy SM, Bloch DA, Mcshane DJ, Fries JF, Cooper NS,
Healey LA, Kaplan SR, Liang MH, Luthra HS. The American Rheumatism
Association 1987 revised criteria for the classification of rheumatoid arthritis.
Arthritis and Rheumatism 1988; 31(3): 315-324.
2. Kirwan JR, Reeback JS. Stanford Health Assessment Questionnaire modified
to assess disability in British patients with rheumatoid arthritis. British Journal
of Rheumatology 1986; 25(2): 206-209
3. Ware JE Jr., Sherbourne CD. The MOS 36-item short-form health survey (SF36). I. Conceptual framework and item selection. Medical Care 1992; 30(6):
473-483
4. Du Montcel ST, Michou L, Petit-Teixeira E, Osorio J, Lemaire I, Lasbleiz S,
Pierlot C, Quillet P, Bardin T, Prum B, Cornelis F, Clerget-Darpoux F. New
classification of HLA-DRB1 alleles supports the shared epitope hypothesis of
rheumatoid arthritis susceptibility. Arthritis and Rheumatism 2005; 52(4): 10631068.
5. Michou L, Croiseau P, Petit-Teixeira E, du Montcel ST,Lemaire I, Pierlot C,
Osorio J, Friqui W, Lasbleiz S, Quillet P, Bardin T, Prum B, Clerget-Darpoux F,
Cornélis F; European Consortium on Rheumatoid Arthritis Families. Validation
of the reshaped shared epitope HLA-DRB1 classification in rheumatoid
arthritis. Arthritis Research & Therapy 2006; 8(3):R79.
104
CHAPTER 3
Serum Matrix Metalloproteinase 3 (MMP-3) in comparison with acute phase
proteins as a marker of disease activity and radiographic damage in early
rheumatoid arthritis
3.1
Introduction
Rheumatoid arthritis (RA) is characterised by a progressive, erosive polyarthritis,
resulting in immune-mediated joint destruction and eventual disability. Components of
the immuno-inflammatory response include acute phase proteins, auto-reactive T
cells, B cells, and their respective inflammatory mediators. The consequence is a
self-perpetuating chronic inflammatory response involving a complex interplay
between infiltrating inflammatory cells and the structural cells of the synovial joint.1,2
Matrix metalloproteinase-3 (MMP-3) is produced predominantly by chondrocytes and
synovial fibroblasts and is found in high concentrations in the synovial fluid, with
elevated levels also found in the serum of RA patients.3 MMP-3 production is
upregulated by the pro-inflammatory cytokines IL-1β, TNF, IFN γ and IL-17A, as well
as by serum amyloid A (SAA), an acute phase reactant, with counter-regulatory
inhibition from IL-4 and IL-13.4-10 Intracellular signalling pathways include NFkB,activating protein 1(AP-1) and STAT pathways which modulate gene
transcription.11 In addition, genetic polymorphisms have also been shown to influence
the level of expression of MMP-3.12,13 Cytokine/SAA-driven production of MMP-3 in
the rheumatoid joint appears to be a key mediator of cartilage destruction, while bone
resorption is facilitated by MMP-3-mediated removal of the outer osteoid layer,
enabling attachment of osteoclasts to the underlying bone.14 Moreover, MMP-3
mediates the proteolytic activation of pro-MMP-9 released from both inflammatory
and structural cells.4 Aside from its role in promoting the recruitment of neutrophils,
monocytes, T cells and osteoclasts,4 MMP-9 promotes the release of membranebound vascular endothelial growth factor (VEGF), thereby contributing to
105
angiogenesis and disease progression.15 With respect to its diagnostic and
prognostic potential, elevated serum concentrations of MMP-3, while having no
specificity for RA, have been found in many,16-24 but not all25-29 studies, to correlate
with disease severity and response to chemotherapy. Serum MMP-3 has been shown
to correlate with histologic severity, and clinical and serological parameters of disease
activity (28TJC,28SJC,CRP and ESR).30,31 Young-Min et al (2007) who evaluated
baseline serum MMP-3 in a cohort of early RA patients, reported an association with
radiographic progression at 2 years and in a follow-up study was still predictive of
radiographic progression at 8.2 years.18,32 Targeted therapy using the combination of
MMP-3 level and DAS28 as outcome measures was associated with the best
results.33
COMP is an extracellular structural glycoprotein important for maintaining cartilage
integrity and facilitating interactions between the extracellular matrix, collagen and
aggrecan.34 Elevated levels reflect cartilage breakdown or increased cartilage
turnover.34 Clinical utility of serum COMP has been limited with respect to its
diagnostic role in RA because of a low sensitivity (15%-48%) and low specificity( 66%
to 69%).35 However, COMP levels may have prognostic value and is possibly
predictive of therapeutic response.34 Elevated levels having been shown to be
associated with progressive disease, with persistently elevated levels correlating with
radiographic progression.36-40 Morozzi et al (2007) reported a potential role of serum
COMP levels in predicting response to anti-TNF with low levels associated with a
more favourable response.39,41
CRP and SAA are synthesised in the liver in response to pro-inflammatory cytokines
such as TNF, IL-1 and IL-6. Monocytes/macrophages, endothelial cells, synovial
fibroblasts and chondrocytes can also produce SAA. Unlike CRP, SAA is also locally
expressed and accumulates in inflamed tissue with histological studies demonstrating
expression of SAA in the perivascular and lining layer of RA synovial tissue, in
regions of leukocyte recruitment and angiogenesis, especially at the cartilage pannus
junction. SAA is apparently a more sensitive marker of inflammation in RA than CRP,
106
with the SAA/CRP ratio, possibly being of more significance.42 SAA augments the
inflammatory response in a cytokine-like fashion by attracting
monocytes/macrophages, leukocytes and T lymphocytes, while promoting neutrophil
survival and endothelial activation, and stimulating the production of the
proinflammatory mediators TNF, IL-1, IL-6, IL-8 and IL-17, thus initiating an
amplifying loop. Exposure of synovial fibroblasts and chondrocytes to SAA promotes
MMP-3 mediated adhesion molecule expression, as well as phagocytosis and
chemotaxis of monocytes and neutrophils, thereby contributing to synovial
inflammation, hyperplasia, angiogenesis and joint destruction.9 SAA has also been
implicated in the pathogenesis of atherosclerosis and premature cardiovascular
disease, an important aspect in the management of patients with RA, thus making it a
potential target for therapy to control both RA and its complications.
The global inter-relationships of MMP-3 and COMP with proven genetic markers of
disease susceptibility, such as the shared epitope (SE), as well as with other
systemic biomarkers such as autoantibodies, cytokines, and the acute phase
reactants CRP and SAA in particular, have not been well characterised.
The objectives of the current study were to assess the relationships between
circulating MMP-3 and: i) SE; ii) biomarkers of inflammation and cartilage
degradation; and iii) disease activity and radiographic changes in comparison with
CRP and SAA in a cohort of predominantly black female South Africans with early
disease-modifying anti-rheumatic drugs (DMARD)-naive RA.
3.2
Patients and Methods (described in detail in chapter 2)
3.2.1 Patients
Baseline data was reviewed from 128 patients who were part of the Gauteng
Rheumatoid Arthritis Trial (GREAT), a prospective study of early DMARD-naive RA
patients.43-45 Patients were recruited from the rheumatology clinics of two tertiary
academic hospitals, Chris Hani Baragwanath Hospital and Steve Biko Academic
107
Hospital, attached to the Universities of the Witwatersrand and Pretoria, South Africa
respectively, and the study was approved by the Research Ethics Committees of the
Faculties of Health Sciences of both institutions.
Demographic data, duration of symptoms, education level, medication history, clinical
assessment including 28 joint count and the presence of extra-articular involvement
were recorded, as was disease activity measured according to the Health
Assessment Questionnaire Disability Index (HAQ-DI) and Simplified Disease Activity
Index (SDAI).43-45
Radiographs of the hands and feet were scored according to the modified Larsen
score. In each case, 32 joint areas were scored: 8 PIP (proximal interphalangeal), 2
thumb IP (interphalangeal), 10 MCP (metacarpophalangeal), 4 areas in each wrist
and 8 MTP (metatarsophalangeal) joints. The maximum score that could be attained
was 160. Radiographs were also assessed for presence or absence of erosive
disease.
3.2.2 Laboratory methods (described in chapter 2)
Venous blood was collected as described previously, followed by prompt separation
of serum and storage at minus 20°C until analysed.
3.2.2.1 Autoantibodies, acute phase reactants, cytokines/chemokines
These were assayed using immunonephelometric, immunofluorimetric, and multiplex
suspension bead array procedures as previously described,43 while haemoglobin
concentrations and erythrocyte sedimentation rates were measured using standard
haematological procedures.
3.2.2.2 MMP-3
Serum concentrations of MMP-3 were measured using the Quantikine® Total MMP-3
Immunoassay (ELISA) according to the instructions supplied by the manufacturer (R
108
& D Systems, Minneapolis, MN, USA) and the results expressed as nanograms
(ng)/ml serum. Serum levels were measured in healthy controls (15 females and 10
males, age range 24-64).
3.2.2.3 Cartilage oligmeric matrix protein (COMP)
Serum levels were measured using the Kamiya Human COMP ELISA system
(Kamiya Biomedical Company, Seattle, WA, USA) and the results expressed as
micrograms (μg)/ml. Serum levels were also measured in 8 of the above control
subjects.
3.2.2.4 Shared epitope (SE) genotyping
This was performed as described previously using high-resolution reverse sequencespecific oligonucleotide probes and Luminex® technology,43 with classification as SE
or non-SE genotypes according to the method of du Montcel et al.46,47
3.2.2.5 Statistical Methods
Descriptive statistics of continuous variables were done by using a measure of central
tendency (mean or median) and a measure of variability (standard deviation or
range). Frequencies and percentages were used to describe categorical variables of
the nominal or ordinal scales. Cytokine data were not normally distributed and
therefore the Spearman’s pairwise correlations were used to find the strengths of
association of each cytokine and demographics with MMP-3 and COMP, and
between themselves. Statistical significance was confirmed by p-values less than
0.05 and corrections for multiple cytokine variables tested with a level of significance
confirmed by p-values of less than 0.003. Statistical analysis was done using STATA
version 12.0 (Stata Corporation, College Station, Texas USA).
109
3.3
Results
3.3.1 Demographic, clinical and laboratory data [autoantibodies, acute phase
reactants, haemoglobin, erythrocyte sedimentation rate (ESR), MMP-3 and
COMP]
As seen in Table 1, the majority of patients were middle-aged black females, with a
mean disease duration of almost 12 months. Patients had high disease activity as
reflected by a mean DAS of 6,28 and moderate- to high- functional disability. 82% of
patients were RF positive and 81% ACPA positive. Erosions were seen in 52% and
the mean Larsen score was 22,4 (±12,7). In patients with disease duration less than
12 months 26.9% of patients had erosive disease.
Serum MMP-3 levels were significantly (p<0.0001) higher in the RA patients relative
to the healthy control subjects. Using cut-off values of 18.6 ng/ml and 1.99 µg/ml for
MMP-3 and COMP respectively (based on the mean values for the healthy control
subjects +2 standard deviations), the mean MMP-3 and COMP values for the control
groups compared favourably with previous studies of 46 and 291 healthy controls
respectively.48,49 The respective frequencies of elevated MMP-3 and COMP levels for
the RA patients were 56.25 % and 34,6 % (Table 1).
3.3.2 Correlations of MMP-3 with clinical indices of disease activity and noncytokine, inflammatory biomarkers
Correlations that achieved statistical significance are shown in Table 2, which
demonstrates associations of serum MMP-3 with SDAI, CRP, SAA, RF and COMP.
There were no statistically significant associations of MMP-3 with HAQ-DI, tender
joint count, nodules, erosions, Larsen score or ACPA.
3.3.3 Correlations of MMP-3 with cytokines/chemokines/growth factors
We have previously reported that the circulating cytokine/chemokine/growth factor
concentrations in this cohort of RA patients are significantly higher than in healthy
control subjects.43 In the current study, circulating MMP-3 was found to correlate
110
positively and significantly with IL-6, IL-8, IL-12, IFN-γ and VEGF (r values 0.27-0.33,
p values <0.003-0.0001), the strongest correlations being observed with IL-8 >IL6>IFN-γ and VEGF (Table 3).
3.3.4 Correlations of COMP with clinical indices of disease activity and noncytokine inflammatory biomarkers
COMP correlated positively and significantly with RF (r = 0.23, p <0.006), but not with
any of the clinical indices or other inflammatory biomarkers. A weak positive
correlation between COMP and smoking history (ever smoked) was also noted (r =
0.17, p <0.04).
3.3.5 Association of MMP-3, SAA, CRP and COMP with SE
Subgroup analysis revealed no differences in MMP-3, SAA, CRP or COMP levels in
risk allele -negative or -positive patients.
3.3.6 Correlations of CRP and SAA with clinical and laboratory indices of
disease activity
Given the key role of SAA in activating the synthesis of MMP-3 by synovial
chondrocytes and fibroblasts,9,50 correlations of this acute phase reactant, as well as
those of CRP, with clinical and traditional non-cytokine indices of disease activity
were determined and these are shown in Table 4. Correlations included evaluation of
the SAA /CRP ratio but this did not reveal any significant associations (data not
shown).Both acute phase proteins, especially CRP, were found to have stronger
associations with disease activity in early RA than MMP-3.
3.4
Discussion
Consistent with data from other studies which reported elevated MMP-3 levels in 6280% of patients,17,51 MMP-3 levels were found to be elevated in 56,25% of patients in
the current study. Although no correlations with SE were detected, MMP-3 was found
to correlate significantly with measures of disease activity, specifically SDAI, ESR,
111
CRP and SAA. In addition to these, correlations with predominantly pro-inflammatory
cytokines, especially IL-8, IL-6, IFN-γ and VEGF (Table 3), were also observed.
Visvanathan et al. in a study of 144 patients with RA showed significant correlations
with IL-8, IL-1β and CRP, the strongest correlation being with CRP (r = 0,601,
p<0,001).52 A smaller study by Ribbens et al in 20 patients with RA showed
correlations of MMP-3 and disease activity score (DAS), CRP, and IL-6 level.23
We are not aware of any studies having examined serum levels of MMP-3 in a
DMARD-naïve early RA cohort of predominantly black females, exploring the
associations with clinical parameters of disease activity, acute phase response, SE,
and a wide range of cytokines, chemokines and growth factors.
Recent developments in the management of RA are aimed at rapid disease control,
as well as early introduction of efficient therapies, which makes the search for useful
biomarkers such as MMP-3 important. This contention is underscored by an interest
in the development of of an automated sero-detection procedure for MMP-3 and the
recent introduction of a commercially available multi-biomarker disease activity
(MBDA) test. The MBDA test includes MMP-3 in the quantitative assessment of
disease activity, having shown significant correlations with DAS 28-CRP
(AUROC=0,77; p<0,001 for sero-positive patients and AUROC=0,70; p<0,001 for
sero-negative patients).53,54
In the current study, the levels of circulating SAA, a mediator of synthesis of MMP-3,
as well as those of CRP, were found to be elevated in 74,4% and 76,1% of patients
respectively, and correlated significantly with measures of disease activity, as
described in previous studies, some of which suggest that SAA is a more sensitive
marker in RA than CRP. However, this was not confirmed in the current study, with
CRP being equivalent or slightly superior to SAA as a marker of disease activity in
early RA. Because SAA is also locally released and expressed in the synovial tissue,
we reasoned that the SAA/CRP ratio may be a more accurate index of disease
activity than the individual biomarkers.42 However, this did not prove to be the case in
112
our cohort. With regard to poor prognostic markers, such as the presence of nodules,
erosive disease and the Larsen score, correlations with CRP and SAA, but not with
MMP-3, were evident. No associations of SAA or CRP with the SE risk alleles or
COMP were observed.
Importantly, the correlations of both CRP and SAA with SDAI were considerably
stronger than those of MMP-3 with SDAI, demonstrating that measurement of either
of these acute phase reactants, but preferably CRP, is probably the best serological
determinant of disease activity in early RA.
Erosive disease was present in 52% of patients but much lower (26.9%) in patients
with disease duration less than 1 year. This compares favourably with early arthritis
studies in different cohorts varying from 15% to 70% (disease duration 1-2 years).21,55
Cartilage destruction is one of the consequences of uncontrolled synovial
proliferation, with serum levels of cartilage degradation products such as COMP
having been shown to be associated with RA disease activity and future radiological
damage.56 In the current study, COMP levels were elevated in 34,6% of patients
compared to other studies showing percentage of elevated COMP levels ranging
from 41 to 67%.41,57 COMP was found to correlate weakly with smoking history (ever
smoked), RF, and MMP-3, but no correlations were found with measures of disease
activity as reflected in some previous studies which described correlations with CRP,
DAS, ESR, and rheumatoid nodules.58 MMP-3 is an important effector of cartilage
metabolism, hence the correlation with COMP is not surprising. Although no
correlation between COMP and radiographic changes were found, this may simply
reflect increased cartilage turnover rather than destruction in early disease.58
Elevated COMP levels may have prognostic value as a predictor of radiographic
progression.40
There were no significant differences in MMP-3, SAA, CRP or COMP levels in the
risk allele negative or positive patients. One would have expected more robust
changes in the risk allele positive group of patients as high-risk SE alleles have been
113
associated with more severe disease.45 However, it appears that no previous studies
have compared levels of these biomarkers in the different risk allele groups according
to the classification used in the current study.
The correlation of MMP-3 with COMP and lack thereof with the acute phase reactants
studied, may suggest that MMP-3 is a better biomarker to predict disease
progression, thus combining measurement of CRP with that of MMP-3 may be a
useful strategy to predict disease progression.28
In conclusion, the results presented in this thesis confirms the observations of
previous studies of an association of either CRP or SAA in combination with MMP-3
and disease activity.21,26 The most significant and original findings of the current study
are: i) elevated levels of circulating MMP-3, which may identify a subset of RA
patients likely to develop severe disease, are significantly associated with SDAI, proinflammatory cytokines, and, most strongly with CRP and SAA; ii) CRP and SAA are
more strongly correlated with disease activity in early RA than MMP-3.
114
Table 1: Demographic, clinical and laboratory data for the group of RA
patients (n = 128)
Parameters
Mean
SD
Age (years)
46,5
12,79
Female (%)
102 (79,6)
Ethnic origin: Black %
119 (92,97)
DAS ESR
6,28
Smoking (%)
28 (21,8)
Nodule (%)
30 (234)
Erosions (%)
64 (52,03)
mHAQ-D1
1,72
0,76
Disease duration(months)
11,70
7,11
SJC
3,51
4,08
ESR
45,02
27,64
RF (IU/ml)
515,83
522 099
aCCP (U/ml)
679,74
626,54
CRP (µg/ml)
26,95
33,95
SAA (ug/ml)
66,00
128.54
Hb (g/l)
12,8
1,76
MMP-3: control group (ng/ml)
7,5
5,4
MMP-3(% elevated)
35,8 (56,3)
43,9
Comp : control group (µg/ml)
1.11
1.85 (34.6)
0.44
0.77
Comp (% elevated)
1,17
DAS: Disease activity score
HDA: High disease activity
SJC: Swollen joint count
RF: Rheumatoid factor
aCCP: anticyclic citrullinated peptide antibodies
CRP: C-reactive protein
SAA : Serum amyloid A
Hb: haemoglobin
ESR: erythrocyte sedimentation rate
MMP : matrix metalloproteinase
Comp: cartilage oligomeric protein
115
Table 2: Correlations of MMP-3 with indices of disease activity and non-cytokine
biomarkers
Parameter
r
p*
n
SDAI
0,29
0,0009
128
CRP
0,39
0,0001
115
SAA
0,40
0,0001
115
COMP
0,21
0,0148
127
RF
0,19
0,0320
128
*p level significance < 0,05
Table 3: Correlations of MMP-3 with cytokines
CYTOKINE
r
p*
n
IL-8
0,33
0,0001
128
IL-6
0,30
0,0005
128
IFNγ
0,28
0,0010
128
VEGF
0,28
0,0014
128
IL-12
0,27
0,0024
128
*p level of significance of < 0,003
116
Table 4: Correlations of SAA and CRP with clinical and laborabory indices of
disease activity
Parameter
SAA*
CRP*
SJC
0,25
0,007
115
0,29
0,002
115
0,54
0,0000
115
0,18
0,047
115
0,83
0,0001
115
0,24
0,009
115
0,21
0,023
115
0,36
0,0001
115
-0,34
0,0002
110
0,26
0,005
110
0,21
0,023
110
0,20
0,044
106
NS
-
0.35
0.0001
115
0.39
0.0001
115
0.63
0.0001
115
0.22
0.014
115
0.25
0.006
115
NS
0.38
0.0001
115
-0.26
0.005
110
0.24
0.01
110
0.19
0.04
110
0.32
0.0006
106
0.28
0.002
115
DAS ESR
SDAI
NODULE
CRP
RF
ACPA
Erythrocyte sedimentation rate
Haemoglobin
Platelets
Erosion
Larsen score
Tender joint count
* For each pair of values the correlation coefficients are uppermost with the
corresponding p and n values underneath.
117
References
1.
Pratt AG, Isaacs JD, Mattey DL. Current concepts in the pathogenesis of early
rheumatoid arthritis. Best Practice & Research Clinical Rheumatology 2009;
23(1):37-48.
2.
Karouzakis E, Neidhart M, Gay RE, Gay S. Molecular and cellular basis of
rheumatoid joint destruction. Immunology Letters 2006; 106(1):8-13.
3.
Kobayashi A, Naito S, Enomoto H, Shiomoi H, Kimura T, Obata K, Inoue K,
Okada Y. Serum levels of matrix metalloproteinase 3 (stromelysin 1) for
monitoring synovitis in rheumatoid arthritis. Archives of Pathology & Laboratory
Medicine 2007; 131(4):563-570.
4.
Jones GJ, Riley GP, Buttle DJ. The role of proteases in pathologies of the
synovial joint. The International Journal of Biochemistry & Cell Biology 2008;
40(6-7):1199-1218.
5.
Galliera E, Banfi G, Corsi MM. Human bone disorders: Pathological role and
diagnostic potential of matrix metalloproteinases. The International Journal of
Biochemistry & Cell Biology 2010; 42(10):1590-1593.
6.
Manicone AM, McGuire JK. Matrix metalloproteinases as modulators of
inflammation. Seminars in Cell & Developmental Biology 2008; 19(1):34-41.
7.
Hueber AJ, Asquith DL, McInnes IB, Miller AM. Embracing novel cytokines in
RA – complexity grows as does opportunity!” Best Practice & Research Clinical
Rheumatology 2010; 24(4):479-487.
8.
Cawston TE, Wilson AJ. Understanding the role of tissue degrading enzymes
and their inhibitors in development and disease. Best Practice & Research
Clinical Rheumatology 2006; 20(5):993-1002.
9.
Connolly M, Mullan RH, McCormick J. Acute-phase serum amyloid A regulates
tumor necrosis factor α and matrix turnover and predicts disease progression in
patients with inflammatory arthritis before and after biologic therapy. Arthritis &
Rheumatism 2012; 64(4):1035-1045.
118
10. Chizzolini C, Brembilla NC, Montanari E, Truchetet ME. Fibrosis and immune
dysregulation is systemic sclerosis. Autoimmunity Reviews 2011; 10(5):276281.
11. Vincenti MP. The matrix metalloproteinase (MMP) and tissue inhibitor of
metalloproteinase (TIMP) genes. Transcriptional and posttranscriptional
regulation, signal transduction and cell-type-specific expression. Methods in
Molecular Biology 2001; 151:121-148.
12. Constantin A, Lauwers-Cancès, Navaux F, Abbal M, van Meerwijk J, Mazières
B, Cambon-Thomsen A, Cantagrel A. Stromelysin 1 (matrix metalloproteinase
3) and HLA-DRB1 gene polymorphisms: Association with severity and
progression of rheumatoid arthritis in a prospective study. Arthritis and
Rheumatism 2002; 46(7):1754-1762.
13. Chen Y, Nixon NB, Dawes PT, Mattey DL. Influence of variations across the
MMP-1 and -3 genes on the serum levels of MMMP-1 and -3 and disease
activity in rheumatoid arthritis. Genes and Immunity 2012; 13(1):29-37.
14. G. Cunnane G, F. Oliver F, C. Beeton C, Cawston TE, Bresnihan B. Early joint
erosions and serum levels of matrix metalloproteinase 1, matrix
metalloproteinase 3, and tissue inhibitor of metalloproteinases 1 in rheumatoid
arthritis. Arthritis & Rheumatism 2001; 44(10):2263-2274.
15. Szekanecz Z, Koch AE. Angiogenesis and its targeting in rheumatoid arthritis.
Vascular Pharmacology 2009; 51(1):1-7.
16. Mamehara A, Sugimoto T, Sugiyama D, Morinobu S, Tsuji G, Kawano S,
Morinobu A, Kumagai S. Serum matrix metalloproteinase-3 as predictor of joint
destruction in rheumatoid arthritis, treated with non-biological disease modifying
anti-rheumatic drugs. The Kobe Journal of Medical Sciences 2010; 56(3):E98107.
17. Ribbens C, Martin y Porras M, Franchimont N, KaiserMJ, Jaspar JM, Damas P,
Houssiau FA, Malaise MG. Increased matrix metalloproteinase-3 serum levels
in rheumatic diseases: relationship with synovitis and steroid treatment. Annals
of the Rheumatic Diseases 2002; 61(2):161-166.
119
18. Young-Min S, Cawston T, Marshall N, Coady D, Christgau S, Saxne T, Robins
S, Griffiths I. Biomarkers predict radiographic progression in early rheumatoid
arthritis and perform well compared with traditional markers. Arthritis &
Rheumatism 2007; 58(10):3236-3247.
19. Mattey DL, Nixon NB, Dawes PT, Ollier WE, Hajeer AH. Association of matrix
metalloproteinase 3 promoter genotype with disease outcome in rheumatoid
arthritis. Genes & Immunity 2004; 5(2):147-149.
20. Tchetverikov I, Lard LR, DeGroot J, VerzijlN, TeKoppele JM, Breedveld FC,
Huizinga TW, Hanemaaijer R. Matrix metalloproteinases-3, -8, -9 as markers of
disease activity and joint damage progression in early rheumatoid arthritis.
Annals of the Rheumatic Diseases 2003; 62(11):1094-1099.
21. Green MJ, Gough AK, Devlin J, Smith J, Astin P, Taylor D, Emery P. Serum
MMP-3 and MMP-1 and progression of joint damage in early rheumatoid
arthritis. Rheumatology [Oxford] 2003; 42(1):83-88.
22. Posthumus MD, Limburg PC, Westra J, van Leeuwen MA, van Rijswijk MH.
Serum matrix metalloproteinase 3 levels during treatment with sulfasalazine or
combination of methotrexate and sulfasalazine in patients with early rheumatoid
arthritis. The Journal of Rheumatology 2002; 29(5):883-889.
23. Ribbens C, Andre B, Jaspar JM, Kaye O, Kaiser MJ, De Groote D, Malaise MG.
Matrix metalloproteinase-3 serum levels are correlated with disease activity and
predict clinical response in rheumatoid arthritis. The Journal of Rheumatology
2000; 27(4):888-893.
24. Posthumus MD, Limburg PC, Westra J, van Leeuwen MA, van Rijswijk MH.
Serum matrix metalloproteinase 3 in early rheumatoid arthritis is correlated with
disease activity and radiological progression. The Journal of Rheumatology
2000; 27(12):2761-2768.
25. Yamanaka H, Matsuda Y, Tanaka M, Sendo W, Nakajima H, Taniguchi A,
Kamatani N. Serum matrix metalloproteinase 3 as a predictor of the degree of
joint destruction during the six months after measurement, in patients with early
rheumatoid arthritis. Arthritis & Rheumatism 2000; 43(4):852-858.
26. Cheung NT, Dawes PT, Poulton KV, Ollier WE, Taylor DJ, Mattey DL. High
serum levels of pro-matrix metalloproteinase-3 are associated with greater
120
radiographic damage and the presence of the shared epitope in patients with
rheumatoid arthritis. The Journal of Rheumatology 2000; 27(4):882-887.
27. Ichikawa Y, Yamada C, Horiki T, hoshina Y, Uchiyama M. Serum matrix
metalloproteinase-3 and fibrin degradation product levels correlate with clinical
disease activity in rheumatoid arthritis. Clinical and Experimental Rheumatology
1998; 16(5):533-540.
28. Young-Min S, Cawston T, Marshall N, Coady D, Christgau S, Saxne T, Robins
S, Griffiths I. Biomarkers predict radiographic progression in early rheumatoid
arthritis and perform well compared with traditional markers. Arthritis &
Rheumatism 2007; 56(10):3236-3247.
29. So A, Chamot M, Peclat V, Gerster JC. Serum MMP-3 in rheumatoid arthritis:
correlation with systemic inflammation but not with erosive status.
Rheumatology 1999; 38(5):407-410.
30. Ma JD, Zhou JJ, Zheng DH, Chen LF, Mo YQ, Wei XN, Yang LJ, Dai L. Serum
matrix metalloproteinase-3 as a noninvasive biomarker of histological synovitis
for diagnosis of rheumatoid arthritis. Mediators of Inflammation 2014;
2014:179284.
31. Ma JD, Ouyang X, Zheng DH, Mo YQ, Zou CJ, Zhu LJ, Li Y, Dai L. Combined
detection of serum matrix metalloproteinase-3 and C-reactive protein in disease
activity measurement in female patients with rheumatoid arthritis. Chinese
Journal of Clinicians (Electronic Edition) 2013; 7(8):3301-3305.
32. Houseman M, Potter C, Marshall N, Lakey R, Cawston T, Griffiths I, Young-Min
S, Isaacs JD. Baseline serum MMP-3 levels in patients with Rheumatoid
Arthritis are still independently predictive of radiographic progression in a
longitudinal observational cohort at 8 years follow up. Arthritis Research &
Therapy 2012; 14(1):R30.
33. Urata Y, Uesato R, Tanaka D, Nakamura Y, Motomura S. Treating to target
matrix metalloproteinase 3 normalisation together with disease activity score
below 2.6 yields better effects than each alone in rheumatoid arthritis patients:
T-4 Study. Annals of the Rheumatic Diseases 2012; 71(4):534-540.
121
34. Tseng S, Reddi AH, Di Cesare PE. Cartilage Oligomeric Matrix Protein (COMP):
A Biomarker of Arthritis. Biomarker Insights 2009; 4:33-44.
35. Nikolaisen C, Rekvig OP, Nossent HC. Diagnostic impact of contemporary
biomarker assays for rheumatoid arthritis. Scandinavian Journal of
Rheumatology 2007; 36(2):97-100.
36. Forslind K, Eberhardt K, Jonsson A, Saxne T. Increased serum concentrations
of cartilage oligomeric matrix protein. A prognostic marker in early rheumatoid
arthritis. British Journal of Rheumatology 1992; 31(9):593-598.
37. Månsson B, Carey D, Alini M, Ionescu M, Rosenberg LC, Poole AR, Heinegård
D, Saxne T. Cartilage and bone metabolism in rheumatoid arthritis. Differences
between rapid and slow progression of disease identified by serum markers of
cartilage metabolism. The Journal of Clinical Investigation 1995; 95(3):10711077.
38. Wolheim FA, Eberhardt KB, Johnson U, Saxne T. HLA DRB1* typing and
cartilage oligomeric matrix protein (COMP) as predictors of joint destruction in
recent-onset rheumatoid arthritis. British Journal of Rheumatology 1997;
36(8):847-849.
39. Morozzi G, Fabbroni M, Bellisai F, Pucci G, Galeazzi M. Cartilage oligomeric
matrix protein level in rheumatid diseases: potential use as a marker for
measuring articular cartilage damage and/or the therapeutic efficacy of
treatments. Annals of the New York Academy of Sciences 2007; 1108:398-407.
40. Andersson ML, Svensson B, Petersson IF, Hafström I, Albertsson K, Forslind K,
Heinegård D, Saxne T. Early increase in serum-COMP is associated with joint
damage progression over the first five years in patients with rheumatoid arthritis.
BMC Musculoskeletal Disorders 2013; 14:229.
41. Morozzi G, Fabbroni M, Bellisai F, Cucini S, Simpatico A, Galeazzi M. Low
serum level of COMP, a cartilage turnover marker, predicts rapid and high
ACR70 response to adalimumab therapy in rheumatoid arthritis. Clinical
Rheumatology 2007; 26(8):1335-1338.
122
42. Kokubun M, Imafuku Y, Okada M, Ohguchi Y, Ashikawa T, Yamada T, Yoshida
H. Serum amyloid A (SAA) concentration varies among rheumatoid arthritis
patients estimated by SAA/CRP ratio. Clinica Chimica Acta 2005; 360(1-2):97102.
43. Meyer PW, Hodkinson B, Ally M, Musenge E, Wadee AA, Fickl H, Tikly M,
Anderson R. Circulating cytokine profiles and their relationships with
autoantibodies, acute phase reactants, and disease activity in patients with
rheumatoid arthritis. Mediators of Inflammation 2010; 2010:158514.
44. Hodkinson B, Musenge E, Ally M, Meyer PW, Anderson R, Tikly M. Response to
traditional disease-modifying anti-rheumatic drugs in indigent South Africans
with early rheumatoid arthritis. Clinical Rheumatology 2012; 31(4):613-619.
45. Meyer PW, Hodkinson B, Ally M, Musenge E, Wadee AA, Fickl H, Tikly M,
Anderson R. HLA-DRB1 shared epitope genotyping using the revised
classification and its association with circulating autoantibodies, acute phase
reactants, cytokines and clinical indices of disease activity in a cohort of South
African rheumatoid arthritis patients. Arthritis Research & Therapy 2011;
13(5):R160.
46. du Montcel ST, Michou L, Petit-Teixeira E, Osorio J, Lemaire I, Lasbleiz S,
Pierlot C, QuilletP,Bardin T, Prum B, Cornelis F, Clerget-Darpoux F. New
classification of HLA-DRB1 alleles supports the shared epitope hypothesis of
rheumatoid arthritis susceptibility. Arthritis & Rheumatism 2005; 52(4):10631068.
47. Courraud PA, Boyer JF, Barnetche T, Abbal M, Cambon-Thomsen A, Cantagrel
A, Constantin A. A new classification of HLA-DRB1 alleles differentiates
predisposing and Protective alleles for rheumatoid arthritis structural severity.
Arthritis & Rheumatism 2006; 54(2):593-599.
48. Tadbir AA, Purshahidi S, Ebrahim H, Khademi B, Malekzadeh M, Mardani M,
Taghva M, Sardari Y. Serum level of MMP-3 in patients with oral squamous cell
carcinoma – lack of association with clinic-pathological features. Asian Pacific
Journal of Cancer Prevention 2012; 13(9):4545-4548.
49. Jordan JM. Cartilage oligomeric matrix protein as a marker of osteoarthritis. The
Journal of Rheumatology. Supplement 2004; 70:45-49.
123
50. Migita K, Kawabe Y, Tominaga M, Origuchi T, Aoyagi T, Eguchi K. Serum
amyloid A protein induces production of matrix metalloproteinases by human
synovial fibroblasts. Laboratory Investigation 1998; 78(5):535-539.
51. Hayato Nagasawa H, Hideto Kameda H, Koichi Amano K, Takeuchi T. Clinical
significance of elevated serum levels of matrix metalloproteinase-3 and Creactive protein in patients with rheumatoid arthritis. APLAR Journal of
Rheumatology 2007; 10(4):295-299.
52. Visvanathan S, Marini JC, Smolen JS, Clair EW, PritchardC, Shergy W, Pendley
C, Baker D, Bala M, Gathany T, Han J, WagnerC. Changes in biomarkers of
inflammation and bone turnover and associations with clinical efficacy following
infliximab plus methotrexate therapy in patients with early rheumatoid arthritis.
The Journal of Rheumatology 2007; 34(7):1465-1474.
53. Curtis JR, Haney D, van der Helm AHM, Shen Y, Knevel R, Cave G, Dirven L.
Validation of a multi-biomarker test for rheumatoid arthritis (RA) disease activity
(Vectra DA) in a multi-cohort study. [Abstract]. Arthritis & Rheumatism 2010;
62(Suppl 10):1782.
54. Hambardzumyan K, Bolce R, Saevarsdottir S, Cruickshank SE, Sasso EH,
Chernoff D, Forslind K, Petersson IF, Geborek P, van Vollenhoven RF.
Pretreatment multi-biomarker disease activity score and radiographic
progression in early RA: results from the SWEFOT trial. Annals of the
Rheumatic Diseases 2014 May 8. [Epub ahead of print]. [doi:
10.1136/annrheumdis-2013-204986].
55. Bugatti S, Manzo A, Caporali R, Montecucco C. Assessment of synovitis to
predict bone erosions in rheumatoid arthritis. Therapeutic Advances in
Musculoskeletal Disease 2012; 4(4):235-244.
56. Fujikawa K, Kawakami A, Tamai M, Uetani M, Takao S, Arima K, Iwamoto N,
Aramaki T, Kawashiri S, Ichinose K, Kamachi M, Nakamura H, Origuchi T, Ida
H, Aoyagi K, Eguchi K. High serum cartilage oligomeric matrix protein
determines the subset of patients with early-stage rheumatoid arthritis with high
serum C-reactive protein, matrix metalloproteinase-3, and MRI-proven bone
erosion. The Journal of Rheumatology 2009; 36(6):1126-1129.
124
57. Gabriella Morozzi G, Marta Fabbroni M, Francesca Bellisai F, Pucci G, Galeazzi
M. Cartilage oligomeric matrix protein level in rheumatic diseases: potential use
as a marker for measuring articular cartilage damage and/or the therapeutic
efficacy of treatments. Annals of the New York Acadademy of Sciences 2007;
1108:398-407.
58. Posey KL, Hecht JT. The role of cartilage oligomeric matrix protein (COMP) in
skeletal disease. Current Drug Targets 2008; 9(10):869-877.
125
CHAPTER 4
Circulating anti-citrullinated peptide antibodies, cytokines, matrix
metalloproteinase-3 (MMP-3), cartilage metabolites and genotypes as
biomarkers of response to disease-modifying anti-rheumatic drug therapy in
early rheumatoid arthritis (Aspects of this chapter have been submitted for
publication to the Journal “Clinical and Experimental Rheumatology” submission code
8199).
4.1
Introduction
Raised levels of anti-citrullinated peptide antibodies (ACPA) have diagnostic and
prognostic value, and have been incorporated into the 2010 Eular/ACR rheumatoid
arthritis (RA) classification criteria.1 Studies investigating therapy-associated
alterations in ACPA levels in patients with early RA have focused predominantly on
biologic disease-modifying anti-rheumatic drugs (DMARDs).2 However, the
association of a decrease in ACPA levels with therapeutic response has been
variable.3-13 On the other hand, raised ACPA levels may account for relapse and
persistence of disease, with the magnitude of the pre-therapy levels being inversely
associated with response to methotrexate (MTX) in early undifferentiated arthritis.14
ACPA levels have not only been shown to correlate with response to anti-TNF
therapy, but are also predictive of response to rituximab.15
The underlying immune-pathogenic mechanisms of RA are complex and involve
numerous well-defined mediators of inflammation culminating in synovial proliferation
and metabolic changes in the joint.
Cytokines play an integral role in the pathogenesis of RA and their importance as
therapeutic targets is well established. However, the utility of serial measurement of
circulating cytokines in RA is not clearly defined. Changes in cytokine levels posttherapy, especially the balance between pro- and anti-inflammatory cytokines, have
the potential to aid in monitoring treatment response, guide future therapy and/or
have prognostic implications.16 For example, a decrease in IL-7 levels after
126
treatment with MTX has been found to correlate with improved clinical measures of
disease activity.17 In addition, TNF levels below 20.1 pg/ml have been shown to be
associated with a good response to MTX, while a low IL-2 level at baseline is an
independent predictor of response to synthetic DMARDs.18 IL-6 levels greater than
4.03 pg/ml post-treatment with MTX have been associated with radiographic
progression.19 The pre-treatment levels of cytokines may also be predictive of
response to biologic DMARDs. Patients with elevated serum TNF levels may require
higher doses of infliximab, while high levels of IL-17 are possibly predictive of a
subgroup of RA patients resistant to TNF blockade.20 Cytokine ratios may also have
prognostic significance, with the IL-6/IL-10 ratio being associated with new coronary
events in the general population.21
Cartilaginous metabolites such as COMP also have the potential to act as biomarkers
in RA management. Pro-inflammatory cytokines increase the production of MMP-3
from chondrocytes facilitating angiogenesis, activation of other proteases and
cartilage and bone destruction.22 Cartilage destruction and increased turnover result
in elevated COMP levels.23 MMP 3 levels have been shown to be associated with
measures of disease activity, progression and functional outcome.24,25 COMP levels
may identify a subset of patients with a poorer prognosis and have been associated
with MRI-proven erosive disease and radiographic progression.26,27
The “shared epitope’’ (SE) is a well-recognized genetic risk factor for, and poor
prognostic marker in RA, being associated with both ACPA positivity and a poorer
response to MTX monotherapy.11,28-31 Patients who do not carry the risk alleles
generally have milder disease, less radiographic progression and are more likely to
respond to DMARDs.
Most studies focused on genotype and profiling of circulating immune biomarkers in
prediction of risk and response to therapy in patients with RA have been undertaken
in developed world countries. However, RA in the developing world, where there is
often little-or-no access to expensive biologic therapies, is associated with as much,
if not more, morbidity, than in developed countries, underscoring the importance of
discerning clinical utility of traditional DMARD-based therapy in limited resource
127
settings. To our knowledge, measurement of the SE/risk allele status and its
association with longitudinal alterations in clinical disease activity, as well as the
concentrations of circulating biomarkers of immune activation, specifically
autoantibodies, acute phase reactants and cytokines/chemokines following initiation
of DMARD-based therapy has not been described in black African patients with early
RA. The clinical data, laboratory findings, treatment and outcome in these patients
were reported by Hodkinson et al (2012).32 The association of the shared epitope with
baselinencirculating autoantibodies,acute phase reactants, cytokines and clinical
indices of activity were reported by Meyer et al (2011).33
Accordingly, the objectives of the present study were: i) to characterize changes in
ACPA and cytokines following synthetic DMARD therapy in an early RA cohort of
predominantly black South African patients; ii) evaluate changes in ACPA and
cytokines in the different genotype and treatment responder groups; and iii) identify
possible baseline biomarkers predictive of disease activity after 6 months of therapy.
4.2
Patients and methods (described in detail in chapter 2)
After obtaining approval from the Research Ethics Committees of the Faculties of
Health Sciences of the University of Pretoria and the University of the Witwatersrand
140 South African patients who met the 1987 revised classification criteria for RA and
were DMARD-naïve, attending two public sector rheumatology clinics, were enrolled
as part of the prospective observational GREAT study.32,33 Informed written consent
was obtained from all participants before recruitment into the study. The majority of
patients were of black ethnicity (n=124, 88.5%), SE positive (n=106 of 115 tested,
92.1%) and sero-positive (RF, n=114, 81.4%; ACPA, n=106 of 126 tested, 84.1%)
with a median (IQR) symptom duration of 9.7 (11.4) months. Thirty four (24.2%)
patients had a history of smoking.
Data collected at baseline and 6 months post therapy included the simple disease
activity index (SDAI),34 rheumatoid factor (RF), ACPA and a panel of cytokines,
chemokines and growth factors. COMP and MMP-3 were collected at baseline only.
128
Subgroup analysis included assessment of SDAI categories and SDAI responder
groups at 6 months. The levels of disease activity were categorised as follows: i)
SDAI ≤ 11 (low disease activity, LDA); ii) SDAI ≤ 26 (moderate disease activity,
MDA); and iii) SDAI > 26 (high disease activity, HDA) (table 1). SDAI response was
categorised according to percentage improvement from baseline SDAI with <50%
being no response, ≥ 50 < 70% a minor response, ≥70 < 85% a moderate response
and ≥85% a major response34 (table 2).
Biomarkers analysed included IL-1β, IL-1Ra, IL-4, IL-5, IL-6, IL-7, IL-8, IL-10, IL-12,
IL-13, IL-17, G-CSF, GM-CSF, IFN-γ, TNF, VEGF, CCL2 ,CCL4,MMP-3 and COMP.
Ratios of IL-1/IL-1Ra, IL 17/ IL 10, IFN- γ/IL- 4, IL-8/IL-10 and IL-6/IL-10 were
calculated. Genotyping was performed by the PCR typing of HLA-DRB1 allele; SE
analysis was done according to the Du Montcel classification, with the groups
pooled according to SE status into risk-allele-positive (S2,S3P) and -negative
(S1,S3D,X) subgroups.35
Patients were followed as per routine standard of care, with the attending clinicians
deciding on the type of DMARD therapy and frequency of follow-up visits. In all, 43
patients were treated with methotrexate (MTX) monotherapy, 33 with MTX in
combination with prednisone; 9 on a combination of MTX and chloroquine (CQ), 1 on
sulfasalazine (SZP), 1 on CQ monotherapy, while the remainder were on varying
combinations of all 3 DMARDs with or without prednisone. The initial dose of MTX
was 7.5-15 mg/week [mean (SD) dose 13.1 (3.2) mg/week], gradually increasing to a
maximum of 25 mg/ week. In 77(55%) patients low-dose prednisone, 5-7.5 mg daily,
was co-prescribed.
4.3
Statistical analysis
The demographic, clinical and biomarker data are described using frequencies and
percentages for categorical types such as gender. Medians and ranges were used to
describe the skewed numerical clinical and biomarker data. The Mann-Whitney test
was used to assess differences in medians for two independent groups, while the
129
Wilcoxon signed-rank test was used to compare two dependent groups (i.e., baseline
vs. six months). The Kruskal-Wallis test was used to determine differences in
medians of more than three independent groups. Statistical significance was
ascertained at the 5% level i.e a p-value of less than 0.05 was deemed significant,
and, where appropriate, Bonferroni corrections to the p-values for multiple tests were
applied. Multivariable analyses were performed using backward stepwise logistic
regression on the outcome disease activity at 6 months and any baseline clinical and
laboratory variables with a p-value ≤ 0.1 in the univariate analysis were included in
the models.
4.4
Results
The baseline demographic, clinical features and laboratory indices of disease activity
for the entire cohort are shown in table 3. After 6 months of DMARD therapy, the
median SDAI declined from 41.39 (IQR 24.1) at baseline to 16 (IQR 15.8) (p=0.001)
(table 4). The categories of response were: i) no response: n=58 (41.4%); ii) minor
response: n=40 (28.5%); iii) moderate response: n=22 (15.7%); and iv) major
response: n=20 (14.1%).There was similarly a significant fall in the median serum
ACPA, several cytokines (IL-4,IL-7, IL-8, G-CSF, VEGF) levels and IL-1β/IL-1Ra and
IL-17/IL-10 ratios (p<0.05).(table 4) Circulating IL-7, IL-8, G-CSF and VEGF levels
were unchanged or increased at 6 months in 41.4%, 30.1%, 43.9% and 42.2% of
patients respectively. However, the magnitudes of the decrements in the
concentrations of these biomarkers did not correlate significantly with the
improvement in SDAI or its individual components of tender and swollen joint counts,
C-reactive protein (CRP), and physician and patient global assessments
ACPA levels decreased in the majority of seropositive cases, 66 (85.7%), and in a
small minority ACPA levels actually increased, 11 (14.2%), while seroconversion and
seroreversion was noted in 3 and 8 patients, respectively, following 6 months of
therapy. There were no associations between alterations in ACPA post-therapy with
either the baseline SDAI or SDAI at 6 months. There were no differences in the levels
of ACPA at baseline or change in ACPA levels after 6 months between patients with
a positive or negative history of ever having smoked.
130
Subgroup analysis of significant changes in SDAI scores and circulating biomarkers,
stratified by type of DMARD therapy, MTX monotherapy vs. the combination of MTX
and prednisone, at 6 months, are shown in table 5. Pre-therapy swollen joint count
(SJC) (p=0.032), CRP (p=0.025) and IL-7 (p=0.011) levels were significantly higher in
the combined treatment subgroup, while the SDAI, IL-8 and VEGF values, although
higher, did not achieve statistical significance. In both subgroups, the SDAI scores,
decreased significantly at 6 months, as did CRP, ACPA and IL-8 levels, but only in
the MTX plus prednisone group was there a significant decrease in IL-7, IL-10 and
VEGF concentrations. The median CRP value post-therapy remained significantly
higher in the combined treatment subgroup relative to the methotrexate monotherapy
group.
Subgroup stratification of data by SDAI categories at 6 months, into LDA, MDA and
HDA, showed that while a decrease in ACPA levels occurred in all 3 categories, the
changes achieved statistical significance in only the LDA and MDA subgroups,
possibly due to a smaller number of patients in the HDA subgroup (table 6). With
respect to differences at baseline, the median ACPA value for the LDA subgroup was
significantly higher than that of the HDA subgroup (p<0.034). In the case of the
cytokines and chemokines, the most notable decreases at 6 months were observed
in the LDA subgroup, with significant decreases in IL-7, IL-8, G-CSF and VEGF
concentrations, while remaining elevated in the HDA subgroup, with the exception of
IL-7. No statistical difference could be shown in steroid use between the 3 SDAI
categories at 6 months. Patients in the LDA subgroup had a statistically significantly
lower SDAI at baseline than the corresponding value for the MDA and HDA
subgroups combined (p=0.007).
Analysis of patients stratified by risk allele status revealed a significantly higher
(p=0.0452) pre-therapy circulating APCA level in the risk allele-positive group, with no
differences between those of the other measured biomarkers, as well as SDAI in the
low- and high-risk allele subgroups.Analysis after 6 months of therapy in this group of
patients stratified by risk allele status, showed that while SDAI scores, CRP and
ACPA concentrations decreased in both the risk allele-negative and positive
131
subgroups, significant falls in the concentrations of several cytokines IL-4, IL-8, IL-10,
G-CSF and VEGF (p=0.0442 – 0.0001) were noted only in the high risk allele
subgroup, however more patients were on steroids in this subgroup (p=0.0368)
(table 7).
On multivariate analysis the changes in the post-therapy concentrations of ACPA, IL7 and G-CSF (p<0.05), may have been influenced by the presence of rheumatoid
nodules Baseline MMP-3 was predictive of a moderate plus disease activity (SDAI
>11) with an OR = 2.8; p<0.032 CI (1.09-7.59) after adjusting for difference in
baseline SDAI. None of the other baseline biomarkers analysed including COMP
were significantly associated with disease activity at 6 months.
4.5 Discussion
Successful therapy with biologic agents in early and established RA has been
associated with decreases in circulating ACPA and pro-inflammatory cytokine levels
though not always correlating with clinical indices of disease activity.2-13 However,
very few studies have addressed this issue in the therapeutic setting of synthetic
DMARDs in early RA, a situation which is likely to be most important in resourcelimited settings, including sub-Saharan Africa
In the present study, ACPA levels decreased significantly post-therapy in the overall
cohort of RA patients, as well as in the various sub-groups categorised according to
type of therapy (MTX monotherapy or MTX in combination with prednisone),
magnitude of baseline SDAI, or risk-allele status. The exception was the HDA
subgroup in which changes in ACPA and CRP did not achieve statistical significance,
possibly due to sustained high disease activity or a smaller number of patients in the
case of ACPA. These findings are in agreement with an earlier study of 66 RA
patients which reported decreases in circulating ACPA in a subgroup of early RA
patients treated with various synthetic DMARDs, but which did not correlate with
either treatment response or type of therapeutic agent.9 In a more recent study, the
absence of a correlation between alterations in ACPA and clinical response following
132
treatment with MTX was also noted in a group of early arthritis patients, although in
contrast to the present study, baseline levels of ACPA were found to be predictive of
response to MTX.12 However, the observation in the current study that the median
pre-therapy ACPA value was highest in the subgroup of patients that achieved LDA,
appears to underscore the lack of a clear relationship between ACPA levels and
response to therapy.
ACPA seroreversion following therapy occurred in only a few patients (n=8) as
reported by others,36 while a total of 3 patients seroconverted. Although ACPA levels
decreased in the majority of patients, an increase was observed in others (n=11,
14.2%). However, no differences in SDAI responses were noted between those
patients with either increased or decreased levels of ACPA post-DMARD therapy.
Pooled analysis of risk allele-positive (S2/S3P) and -negative (S1/S3D/X) patients
revealed a significantly lower baseline ACPA titre in the latter group. Previous studies
have described higher circulating concentrations of ACPA in RA patients positive for
HLA-DRB*0104 relative to those in patients positive for HLA-DRB*0103, while the S2
and S3P risk alleles have also been reported to be associated with increased levels
of ACPA.37-39
The association between smoking and predisposition for development of seropositive
RA is well recognised.40 However, baseline ACPA levels, as well as the magnitudes
of the post-therapy decrements thereof, did not differ significantly between smokers
and non-smokers in the present study. ACPA and smoking status may be influenced
by ethnicity as a lack of association shown by Mikuls et al. in an African American
cohort.41
Taken together, the aforementioned findings clearly document significant decreases
in circulating ACPA concentrations following 6 months of therapy with synthetic
DMARDs, mostly MTX, with or without prednisone, irrespective of baseline SDAI,
shared epitope or smoking status. However, the lack of a compelling correlation with
changes in clinical indices of disease activity appears to exclude the utility of serial
measurement of ACPA as a strategy to monitor the efficacy of therapy with these
133
agents over the initial 6 month period. In contrast, other studies based on therapy
with biologic DMARDs such as adalimumab, rituximab and infliximab reported that
measurement of ACPA has the potential to guide therapy with these agents.42-45
Importantly, and not addressed in the present study, ACPA status, as well as
alterations in ACPA during the course of RA may have prognostic value as these
have been associated with both disease severity and radiographic progression.46-55
Moreover, ACPA levels have also been found to correlate with diastolic dysfunction
and reduced myocardial mass in patients with RA.56,57
With respect to pro-inflammatory cytokines/chemokines, these generally declined
significantly in the entire cohort of patients, as did the IL-1β/IL-1Ra and IL-17/IL-10
ratios. However, as reported by others alterations in these parameters were not
significantly correlated with improvements in clinical indices of disease activity
following treatment of RA patients for 6 month with the combination of MTX and
prednisone.58 Increases in the levels of IL-8 following treatment with MTX, which
were noted in 30% of patients in the present study, have been shown to be
associated with accelerated radiographic progression.59
With respect to the subgroup analyses comparison between the MTX monotherapy
and MTX + prednisone subgroups revealed significant decreases in IL-7, IL-8, IL-10,
and VEGF in the latter subgroup, but only IL-8 in the former, possibly supporting the
role of low-dose corticosteroids in the management of early RA. Interestingly pretherapy IL-7 and CRP levels, which were unknown to the attending clinicians, were
significantly higher in those patients earmarked to receive combined therapy, possibly
consistent with a perception of more severe disease, perhaps influenced by the
increased SJC (p=0.032) noted in the patients who received MTX and prednisone.
In the various subgroups categorised according to the magnitude of the SDAI at 6
months, the concentrations of IL 4, IL-7, IL-8, IL-10, G-CSF and VEGF decreased
significantly in the LDA subgroup post therapy, while IL-8 and VEGF, and only IL-7,
declined in the MDA and HDA subgroups, respectively. Persistently elevated levels of
IL-8, G-CSF and VEGF in the HDA subgroup may implicate neutrophilic inflammation
in the immunopathogenesis of more severe disease. However, the lack of correlation
134
between circulating concentrations of the individual cytokines with therapy-associated
alterations in disease activity appears to exclude both baseline and serial
measurement thereof as a strategy to monitor the success of treatment with synthetic
DMARDs. Again, this contention contrasts with the apparent utility of baseline
measurement of cytokines such as IL-6, IL-8, CCL2 and/or TNF as predictors of
response to biologic DMARDs, including golimumab, etanercept and infliximab.16,60-64
Differences between cytokine profiles in the risk allele-negative and –positive
subgroups were also detected post-therapy, possibly reflecting different inflammatory
mechanisms and responses to therapy in relation to genotype, although significantly
more patients were on steroids in the risk allele subgroup.
The current paradigm of treating RA is to achieve a low disease activity level i.e SDAI
< 11 or preferably remission (SDAI<3.3) by 6 months of therapy. Biomarkers that
could identify a group of patients with a poorer outcome may allow for cost-effective
stratification and intensive management of patients. However, in this study the only
baseline marker of those analysed, associated with disease activity (SDAI > 11) after
6 months of therapy, was MMP-3. Studies have reported an association of baseline
MMP-3 with disease activity and/or radiographic progression mostly at 1 year and
beyond,24,65,66 but not many studies have reported on the utility of baseline MMP-3
levels and disease activity as early as 6 months after initiating therapy.
Potential limitations of the study include firstly, the fairly small numbers of patients in
some of the subgroups, especially the HDA subgroup, while the presence of
nodulosis and lack of treatment randomization may have influenced the analyses.
Secondly, 6 months might be too short a period in which to detect associations of
circulating biomarkers of disease activity with clinical responses to
therapy.Conversely, the strength of the study in our view is that we were able to
characterize clinical indices of disease activity and systemic biomarker profiles preand post-therapy with synthetic DMARD therapy in patients with relatively early
disease, with the potential to guide future therapy. Notwithstanding the apparent lack
of clinical utility of serial monitoring of ACPA and pro-inflammatory cytokines to
monitor responses to synthetic DMARD therapy, some of the observations are
135
noteworthy. These include: i) that HDA at baseline is associated with a poor clinical
response to therapy in the setting of persistently elevated levels of VEGF, IL-8 and GCSF, underscoring the possible need for more intensive management to optimize
patient outcome; ii) combining MTX with oral corticosteroids is associated with a more
robust decline in cytokine levels; iii) ACPA levels measured pre-therapy were found to
be significantly higher in patients with the risk allele genotype; and iv) MMP-3 at
baseline may be able to identify a sub-group of patients requiring more intensive
management.
4.6
Conclusions
These findings show that synthetic DMARD therapy for 6 months is associated with a
significant decrease in ACPA and pro-inflammatory cytokines, in tandem with
therapeutic response. The lack of correlation with measures of disease activity
precludes serial measurement of these biomarkers as a strategy to monitor early
responses to therapy with synthetic DMARDs. Nonetheless, the long-term prognostic
value of these biomarkers, especially in relation to radiographic progression and
functional disability, remains to be established, as does their potential to guide future
cost effective therapies, of particular importance in resource-limited settings.
Table 1: SDAI catagories
Remission
≤3
LDA
>3≤11
MDA
>11 ≤ 26
HAD
>26
Table 2: SDAI measures of response
None
<50%
Minor
<70%
Moderate
<85%
Major
≥85%
136
Table 3: Demographics of patients (n=140).
n or
Median
124.0
110.0
47.8
9.7
34.0
27
114.0
106.0
106.0
17.1
41.4
Black ethnicity (%)
Females (%)
Age in years, median (IQR)
Symptom duration in months, median (IQR)
Smoking history (%)
Nodulosis (%)
Rheumatoid factor positive (%)
ACPA [n=126] (%)
Shared Epitope positive [n=115] (%)
CRP baseline,median (IQR)
SDAI baseline, median (IQR)
% or IQR
88.5
79.2
14.4
11.4
24.2
19.3
81.4
84.1
92.1
42.3
24.1
Table 4: Significant changes in SDAI scores and circulating biomarker concentrations
following 6 months DMARD therapy.
Baseline
6 Months
Variable
n
p value
Median
IQR
Median
IQR
SDAI
CRP (μg/ml)
ACPA (IU/ml)
IL-4 (pg/ml)
IL-7 (pg/ml)
IL-8 (pg/ml)
G-CSF (pg/ml)
VEGF (pg/ml)
Ratio IL-1 / IL-1ra
Ratio IL-17 / IL-10
140
140
100
32
123
123
123
123
37
30
41.39
17.10
516.60
4.70
20.32
8.79
13.82
168.56
0.05
10.42
24.10
42.25
1027.75
10.28
82.22
12.65
60.02
441.33
0.05
15.61
16 .00
8.90
255.65
1.51
16.69
5.70
7.23
92.62
0.03
3.72
15.81
17.35
677.20
6.53
23.46
7.09
51.61
167.21
0.06
3.81
0.0001
0.0001
0.0001
0.0075
0.0013
0.0001
0.0083
0.001
0.013
0.0001
Only those biomarkers which decreased significantly post-therapy are shown. (Changes in
whole cohort shown in table 8)
137
Table 5: Subgroup analysis of changes in SDAI values and circulating biomarkers
following 6 months DMARD, for the methotrexate (MTX) monotherapy and
methotrexate- prednisone combination therapy groups.
Variable
MTX Only
Baseline
n
SDAI
CRP (μg/ml)
ACPA (IU/ml)
IL-7(pg/ml)
IL-8 (pg/ml)
IL-10(pg/ml)
VEGF(pg/ml)
Variable
MTX PRED
Median
43
43
36
42
42
7
42
24.75
21.30
844.50
64.04
8.84
48.16
240.82
Baseline
n
SDAI
CRP (μg/ml)
ACPA (IU/ml)
IL-7 (pg/ml)
IL-8 (pg/ml)
IL-10 (pg/ml)
VEGF (pg/ml)
33.58
9.90
501.60
14.78
7.68
6.68
114.16
IQR
Median
33
33
29
28
28
14
28
IQR
38.90
20.64
35.90
36.50
598.90 1036.60
46.67
161.13
10.35
13.27
12.99
18.55
318.91
482.44
6 Months
Median
16.03
6.90
310.90
14.17
5.01
8.41
97.52
IQR
11.05
13.40
559.60
19.73
5.32
23.41
184.54
6 Months
Median
18.00
11.60
367.20
21.46
5.84
8.50
77.08
IQR
18.73
19.10
699.50
31.06
4.03
23.95
159.38
p value
0.0001
0.0048
0.0033
NS
0.0091
NS
NS
p value
0.0001
0.0005
0.0001
0.0026
0.0002
0.048
0.0044
Table 6: Significant changes in circulating biomarker concentrations, stratified
according to SDAI at 6 months.
SDAI Category ≤ 11
Baseline
Variable
SDAI
CRP (μg/ml)
ACPA (IU/ml)
IL-4 (pg/ml)
IL-7 (pg/ml)
IL-8 (pg/ml)
IL-10 (pg/ml)
G-CSF (pg/ml)
VEGF (pg/ml)
n
Median
43
43
30
9
36
36
9
36
36
IQR
32.42
24.71
9.50
20.80
936.55 1162.70
7.50
7.30
22.22
82.20
8.05
11.48
13.30
15.25
21.99
99.56
154.96
442.84
6 Months
Median
6.40
6.90
344.60
1.50
16.19
5.56
6.85
5.78
98.62
IQR
5.34
8.10
671.80
4.00
16.74
6.15
5.64
55.94
158.33
p value
0.0001
0.0054
0.0005
0.028
0.005
0.0169
0.0382
0.0095
0.0036
138
SDAI Category > 11 ≤ 26
Variable
Baseline
n
SDAI
CRP (μg/ml)
ACPA (IU/ml)
IL-4 (pg/ml)
IL-7 (pg/ml)
IL-8 (pg/ml)
IL-10 (pg/ml)
G-CSF (pg/ml)
VEGF (pg/ml)
Median
62
62
42
14
55
55
14
55
55
IQR
42.25
23.39
26.50
45.60
483.35 1025.50
4.08
13.73
19.18
61.68
8.94
8.92
14.07
39.13
9.45
42.11
168.10
420.05
6 Months
Median
17.15
8.90
252.15
2.53
18.17
5.85
19.18
7.23
84.30
IQR
6.86
17.00
564.70
8.47
26.61
6.99
19.91
51.61
169.40
p value
0.0001
0.0001
0.0001
NS
NS
0.0029
NS
NS
0.0102
SDAI Category >26
Variable
SDAI
CRP (μg/ml)
ACPA (IU/ml)
IL-4 (pg/ml)
IL-7 (pg/ml)
IL-8 (pg/ml)
IL-10 (pg/ml)
G-CSF (pg/ml)
VEGF (pg/ml)
Baseline
n
Median
35
35
28
9
32
32
9
32
32
43.00
13.00
353.60
1.24
46.67
8.55
5.76
11.7
170.78
IQR
28.50
44.30
917.00
7.18
113.17
27.69
8.93
39.12
484.05
6 Months
Median
36.21
17.00
177.00
0.62
13.66
6.37
5.81
9.87
126.02
IQR
11.91
24.40
714.40
1.0
32.18
14.49
2.97
54.15
187.48
p value
NS
NS
NS
NS
0.0301
NS
NS
NS
NS
139
Table 7: Changes in SDAI scores and circulating biomarkers, stratified by shared
epitope (risk allele) status.
Risk allele negative
Baseline
Variable
n
SDAI
CRP (μg/ml)
ACPA (IU/ml)
IL-4 (pg/ml)
IL-8 (pg/ml)
IL-10 (pg/ml)
G-CSF (pg/ml)
VEGF (pg/ml)
Median
47
47
40
10
44
10
44
44
IQR
39.18
23.50
18.00
43.60
331.90 1006.60
2.55
7.99
7.68
14.15
7.34
27.79
14.42
36.94
130.52
512.22
6 Months
Median
18.00
7.00
185.60
5.24
5.23
19.18
12.18
92.22
IQR
19.50
14.60
654.15
7.18
5.98
19.91
81.23
148.07
p value
0.0001
0.0002
0.0032
NS
NS
NS
NS
NS
Risk allele positive
Variable
SDAI
CRP (μg/ml)
ACPA (IU/ml)
IL-4 (pg/ml)
IL-8 (pg/ml)
IL-10 (pg/ml)
G-CSF (pg/ml)
VEGF (pg/ml)
Baseline
n
Median
68
68
56
19
65
19
65
65
40.04
17.50
561.10
5.00
9.49
11.24
8.66
190.43
IQR
22.12
38.70
911.05
9.80
12.15
18.78
42.11
401.81
6 Months
Median
16.00
10.10
277.60
1.00
5.97
5.53
1.85
95.76
IQR
16.52
17.50
677.80
1.50
6.20
6.52
24.74
160.99
p value
0.0001
0.0005
0.0001
0.0007
0.0001
0.044
0.0442
0.0056
140
Table 8: Changes in SDAI and circulating biomarker concentrations following 6 months of
sDMARD therapy
p
Variable
N
Median
min
max
iqr
value
*
SDAI b
140
41.39
5.28
77
23.67
SDAI 6**
140
16
0.1
69.77
15.805
0.0001
CRP b
140
17.1
1
303
42.25
CRP 6
140
8.9
1
83.9
17.35
0.0001
ACPA b
100
516.6
1.9
2526.8
1027.75
ACPA 6
100
255.65
1.1
1420.3
677.2
0.0001
IL-1 β b
51
6.23
0.12
191.32
13.69
IL-1 β 6
51
3.8
0.35
199.74
18.17
NS
IL-1Ra b
123
80.06
0
5012.27
288.51
IL-1Ra 6
123
80.54
0
12084.64
427.81
NS
IL-2 b
32
0
0
216.91
7.41
IL-2 6
32
0
0
267.6
13.88
NS
IL-4 b
32
4.7
0
904.79
10.28
IL-4 6
32
1.42
0
62.61
5.31
0.0075
IL-6 b
123
24.35
0
791.93
67.93
IL-6 6
123
14.23
0
778.82
51.89
NS
IL-7 b
123
20.32
0
6825.1
82.22
IL-7 6
123
16.69
0
3596
23.46
0.0013
IL-8 b
123
8.79
0
36698.94
12.65
IL-8 6
123
5.7
0
1618.59
7.09
0.0001
IL-10 b
32
11.435
0
1172.06
29.09
IL-10 6
32
8.145
1.8
1111
21.71
NS
IL-12 b
123
11.93
0.63
3557.84
64.8
IL-12 6
123
17.75
0.41
8144.25
74.79
NS
IL-17 b
32
0
0
23.9
0
IL-17 6
32
0
0
13.87
0
NS
G-CSF b
123
13.82
0
4817.44
60.02
G-CSF 6
123
7.23
0
1328.97
51.61
0.0083
GM-CSF b
123
1.08
0
1458.59
44.37
GM-CSF 6
123
0
0
2571.87
82.37
NS
IFNγ b
123
42.64
0
11151.66
264.72
IFNγ 6
123
27.19
0
6394.31
444.72
NS
TNF b
123
11.67
0.55
2951.68
69.55
TNF 6
123
12.15
1.33
3107.1
93.82
NS
VEGF b
123
168.56
0
4503.15
441.33
VEGF 6
123
92.62
0
5007.35
167.21
0.001
CCl-2 b
32
10.65
0
521.48
100.955
CCl-2 6
32
50.65
0
566.52
81.59
NS
CCl-4 b
32
100.905
26.67
344.72
91.10001
CCl-4 6
32
96.275
27.26
326.37
62.46
NS
141
IL-1/IL1ra b
IL-1/IL1ra 6
IL-17/IL10 b
IL-17/ IL10 6
IFNg/ IL-4
b
IFNg/ IL-4
6
IL-8/ IL10 b
IL-8/ IL10 6
IL-6/ IL10 b
IL-6/ IL10 6
37
0.050012
0.0057609
0.9042904
0.0487324
37
0.0282269
0.0029457
0.4552684
0.0622231
30
10.41581
0.3762998
118.3059
15.60833
30
3.723691
0.6393507
22.96679
5.373202
24
25.17977
0
179.2
28.12688
24
21.07841
1.105882
83.55289
33.04997
30
0.8005226
0.0097521
45.19495
2.059342
30
0.796825
0.0068947
27.28358
1.268058
30
1.046355
0
33.66154
2.474186
30
1.834637
0
37.93333
6.346017
0.013
0.0001
NS
NS
NS
*b – baseline
*6 – 6 months
*NS – not significant
142
References
1.
Aletaha D, Neogi T, Silman AJ, FunovitsJ, Felson DT, Bingham CO 3rd,
Birnbaum NS, Burmester GR, Bykerk VP, Cohen MD, Combe B, Costenbader
KH, Dougados M, Emery P, Ferraccioli G, Hazes JM, Hobbs K, Huizinga TW,
Kavanaugh A, Kay J, Kvien TK, Laing T, Mease P, Ménard HA, Moreland LW,
Naden RL, Pincus T, Smolen JS, Stanislawska-Biernat E, Symmons D, Tak PP,
Upchurch KS, Vencovský J, Wolfe F, Hawker G.. 2010 Rheumatoid arthritis
classification criteria: an American College of Rheumatology/European League
Against Rheumatism collaborative initiative. Arthritis and Rheumatism 2010;
62(9):2569-2581.
2.
Modi S, Soejima M, Levesque MC. The effect of targeted rheumatoid arthritis
therapies on anti-citrullinated protein autoantibody levels and B cell responses.
Clinical and Experimental Immunology 2013; 173(1):8-17.
3.
Morozzi G, Fabbroni M, Bellisai F, Cucini S, Simpatico A, Galeazzi M. Low
serum level of COMP, a cartilage turnover marker, predicts rapid and high
ACR70 response to adalimumab therapy in rheumatoid arthritis. Clinical
Rheumatology 2007; 26(8):1335-1338.
4.
Alex P, Szodoray P, Knowlton N, Dozmorov IM, Turner M, Frank MB, Arthur RE,
Willis L, Flinn D, Hynd RF, Carson C, Kumar A, El-Gabalawy HS, Centola M.
Multiplex serum cytokine monitoring as a prognostic tool in rheumatoid arthritis.
Clinical and Experimental Rheumatology 2007; 25(4):584-592.
5.
Alessandri C, Bombardieri M, Papa N, Cinguini M, Magrini L, Tincani A, Valesini
G. Decrease of anti-cyclic citrullinated peptide antibodies and rheumatoid factor
following anti-TNFalpha therapy (infliximab) in rheumatoid arthritis is associated
with clinical improvement. Annals of the Rheumatic Diseases 2004;
63(10):1218-1221.
143
6.
Bobbio-Pallavicini F, Alpini C, Caporali R, Avalle S, Bugatti S, Montecucco C.
Autoantibody profile in rheumatoid arthritis during long-term infliximab treatment.
Arthritis Research & Therapy 2004; 6(3):R264–R272.
7.
Caramaschi P, Biasi D, Tonolli E, Pieropan S, Martinelli N, Carletto A, Volpe A,
Bambara LM. Antibodies against cyclic citrullinated peptides in patients affected
by rheumatoid arthritis before and after infliximab treatment. Rheumatology
International 2005; 26(1):58-62.
8.
Chen HA, Lin KC, Chen CH, Liao HT, Wang HP, Chang HN, Tsai CY, Chou CT.
The effect of etanercept on anti-cyclic citrullinated peptide antibodies and
rheumatoid factor in patients with rheumatoid arthritis. Annals of the Rheumatic
Diseases 2006; 65(1):35-39.
9.
Mikuls TR, O’Dell JR, Stoner JA, Parrish LA, Arend WP, Norris JM, Holers VM.
Association of rheumatoid arthritis treatment response and disease duration
with declines in serum levels of IgM rheumatoid factor and anti-cyclic
citrullinated peptide antibody. Arthritis and Rheumatism 2004; 50(12):37763782.
10. Atzeni F, Sarzi-Puttini P, Dell' Acqua D, de Portu S, Cecchini G, Cruini C,
Carrabba M, Meroni PL. Adalimumab clinical efficacy is associated with
rheumatoid factor and anti-cyclic citrullinated peptide antibody titer reduction: a
one-year prospective study. Arthritis Research & Therapy 2006; 8(1):R3.
11. Braun J, Rau R. An update on methotrexate. Current Opinion in Rheumatology
2009; 21(3):216-223.
12. Bobbio-Pallavicini F, Caporali R, Bugatti S, Montecucco C. What can we learn
from treatment-induced changes in rheumatoid factor and anti-citrullinated
Peptide antibodies? The Journal of Rheumatology 2008; 35(10):1903-1905.
144
13. De Rycke L, Verhelst X, Kruithof E, Van den Bosch F, Hoffman IE, Veys
EM,DeKeyser F. Rheumatoid factor, but not anti-cyclic citrullinated peptide
antibodies, is modulated by infliximab treatment in rheumatoid arthritis. Annals
of the Rheumatic Diseases 2005; 64(2):299–302.
14. Visser K, Verpoort KN, van Dongen H, van der Kooij SM, Allaart CF, Toes RE,
HuizingaTW, van der Helm-van Mil AH. Pretreatment serum levels of anti-cyclic
citrullinated peptide antibodies are associated with the response to
methotrexate in recent-onset arthritis. Annals of the Rheumatic Diseases 2008;
67(8):1194-1195.
15. Lal P, Su Z, Holweg CT, Silverman GJ,Schwartzman S, Kelman A, Read S,
Spaniolo G, Monroe JG, Behrens TW, Townsend MJ. Inflammation and
autoantibody markers identify rheumatoid arthritis patients with enhanced
clinical benefit following rituzimab treatment. Arthritis and Rheumatism 2011;
63(12):3681-3691.
16. Kasama T, Isojima S, Umemura M, Tsukamoto H, TokunagaT, Furuya H, Yanai
R,Takahashi R, Nakamura M, Inagaki K. Serum macrophage migration
inhibitory factor levels are correlated with response to tocilizumab therapy in
patients with rheumatoid arthritis. Rheumatology International 2014; 34(3):429433.
17. van Roon JA, Jacobs K, Verstappen S, Bijlsma J, Lafeber F. Reduction of
serum interleukin 7 levels upon methotrexate therapy in early rheumatoid
arthritis correlates with disease suppression. Annals of the Rheumatic Diseases
2008; 67(7):1054-1055.
18. Maillefert JF, Puéchal X, Falgarone G, Lizard G, Ornetti P, Solau E, Legré V,
Lioté F, Sibilia J, Morel J, Maynadié M; Réseau Rhumato Study Group.
145
Prediction of response to disease modifying antirheumatic drugs in rheumatoid
arthritis. Joint Bone Spine 2010, 77(6):558-563.
19. Nishina N, Kaneko Y, Kameda H, Kuwana M, Takeuchi T. Reduction of plasma
IL-6 but not TNF-α by methotrexate in patients with early rheumatoid arthritis: a
potential biomarker for radiographic progression. Clinical Rheumatology 2013;
32(11):1661-1666.
20. Simsek I. Predictors of response to TNF inhibitors in rheumatoid arthritis – do
we have new tools for personalized medicine? Bulletin of the NYU Hospital for
Joint Diseases 2012; 70(3):187-190.
21. Kilic T, Ural D, Ural E, Yumuk Z, Agacdiken A, Sahin T, Kahraman G, Kozdag
G, Vural A, Komsuoglu B.. Relation between proinflammatory to antiinflammatory cytokine ratios and long-term prognosis in patients with non-ST
elevation acute coronary syndrome. Heart 2006; 92(8):1041-1046.
22. Manicone AM, McGuire JK. Matrix metalloproteinases as modulators of
inflammation. Seminars in Cell & Developmental Biology 2008; 19(1): 34-41.
23. Turesson C, Bergström U, Jacobsson LT, Truedsson L, Berglund G, Saxne T.
Increased cartilage turnover and circulating autoantibodies in different subsets
before the clinical onset of rheumatoid arthritis. Annals of the Rheumatic
Diseases 2011; 70(3):520-522.
24. Galil SM, El-Shafey AM, Hagrass HA, Fawzy F, Sammak AE. Baseline serum
level of matrix metalloproteinase-3 as a biomarker of progressive joint damage
in rheumatoid arthritis patients. International Journal of Rheumatic Diseases
2014 Oct 7. doi: 10.1111/1756-185X.12434. [Epub ahead of print].
25. Shinozaki M1, Inoue E, Nakajima A, Hara M, Tomatsu T, Kamatani N,
Yamanaka H. Elevation of serum matrix metalloproteinase-3 as a predictive
146
marker for the long-term disability of rheumatoid arthritis patients in a
prospective observational cohort IORRA. Modern Rheumatology 2007; 17(5):
403-408.
26. Fujikawa K, Kawakami A, Tamai M, Uetani M, Takao S, Arima K, Iwamoto N,
Aramaki T, Kawashiri S, Ichinose K, Kamachi M, Nakamura H, Origuchi T, Ida
H, Aoyagi K, Eguchi K. High serum cartilage oligomeric matrix protein
determines the subset of patients with early-stage rheumatoid arthritis with high
serum C-reactive protein, matrix metalloproteinase-3, and MRI-proven bone
erosion. The Journal of Rheumatology 2009; 36(6): 1126-1129.
27. Skoumal M, Kolarz G, Klingler A. Serum levels of cartilage oligomeric matrix
protein. A predicting factor and a valuable parameter for disease management
in rheumatoid arthritis. Scandinavian Journal of Rheumatology 2003; 32(3) :15661.
28. Niewold TB, Harrison MJ, Paget SA. Anti-CCP antibody testing as a diagnostic
and prognostic tool in rheumatoid arthritis. QJM: monthly journal of the
Association of Physicians 2007; 100(4):193-201.
29. Haroon N, Misra R, Aggarwal A. Tailor-made therapy in rheumathoid arthritis:
fact or fiction? The Israel Medical Association Journal: IMAJ 2008; 10(2):139141.
30. Halilova KI, Brown EE, Morgan SL, Bridges SLJr., Hwang MH, Arnett DK,
Danila MI. Markers of treatment response to methotrexate in rheumatoid
arthritis: where do we stand? International Journal of Rheumatology 2012;
2012: Article ID 978396.
147
31. Romão VC, Canhão H, Fonseca JE. Old drugs, old problems: where do we
stand in prediction of rheumatoid arthritis responsiveness to methotrexate and
other synthetic DMARDs? BMC Medicine 2013; 11:17.
32. Hodkinson B, Musenge E, Ally M, Meyer PW, Anderson R, Tikly M. Response to
traditional disease-modifying anti-rheumatic drugs in indigent South Africans
with early rheumatoid arthritis. Clinical Rheumatology 2012; 31(4):613-619.
33. Meyer PW, Hodkinson B, Ally M, Musenge E, Wadee AA, Fickl H, Tikly M,
Anderson R. Circulating cytokine profiles and their relationships with
autoantibodies, acute phase reactants, and disease activity in patients with
rheumatoid arthritis. Mediators of Inflammation 2010; 2010: Article ID 158514.
34. Aletaha D, Martinez-Avila J, Kvien TK, Smolen JS. Definition of treatment
response in rheumatoid arthritis based on the simplified and the clinical disease
activity index. Annals of the Rheumatic Diseases 2012; 71(7):1190-1196.
35. Michou L, Croiseau P, Petit-Teixeira E, du Montcel ST, Lemaire I, Pierlot C,
Osorio J, Frigui W, Lasbleiz S, Quillet P, Bardin T, Prum B, Clerget-Darpoux F,
Cornélis F; European Consortium on Rheumatoid Arthritis Families. Validation
of the reshaped shared epitope HLA-DRB1 classification in rheumatoid arthritis.
Arthritis Research & Therapy 2006; 8(3):R79.
36. Kastbom A, Strandberg G, Lindroos A, Skogh T. Anti-CCP antibody test predicts
the disease course during 3 years in early rheumatoid arthritis (the Swedish
TIRA project). Annals of the Rheumatic Diseases 2004; 63(9):1085-1089.
37. Kurkó J, Besenyei T, Laki J, Glant TT, Mikecz K, Szekanecz Z. Genetics of
rheumatoid arthritis — a comprehensive review. Clinical Reviews in Allergy &
Immunology 2013; 45(2):170–179.
148
38. Kapitány A, Szabó Z, Lakos G, Aleksza M, Végvári A, Soós L. Karányi Z, Sipka
S, Szegedi G, Szekanecz Z.. Associations between serum anti-CCP antibody,
rheumatoid factor levels and HLA-DR4 expression in Hungarian patients with
rheumatoid arthritis. The Israel Medical Association Journal: IMAJ 2008;
10(1):32-36.
39. Irigoyen P, Lee AT, Wener MH, Li W, Kern M, Batliwalla F, Lum RF, Massarotti
E, Weisman M, Bombardier C, Remmers EF, Kastner DL, Seldin MF, Criswell
LA, Gregersen PK. Regulation of anti-cyclic citrullinated peptide antibodies in
rheumatoid arthritis: contrasting effects of HLA-DR3 and the shared epitope
alleles. Arthritis and Rheumatism 2005; 52(12):3813-3818.
40. Lee DM, Phillips R, Hagan EM, Chibnik LB, Costenbader KH, Schur PH.
Quantifying anti-cyclic citrullinated peptide titres: clinical utility and association
with tobacco exposure in patients with rheumatoid arthritis. Annals of the
Rheumatic Diseases 2009; 68(2):201-208.
41. Mikuls TR, Hughes LB, Westfall AO, Holers VM, Parrish L, van der Heijde D,
van Everdingen M, Alarcón GS, Conn DL, Jonas B, Callahan LF, Smith EA,
Gilkeson G, Howard G, Moreland LW, Bridges SL Jr. Cigarette smoking,
disease severity and autoantibody expression in African Americans with recentonset rheumatoid arthritis. Annals of the Rheumatic Diseases 2008;
67(11):1529-1534.
42. da Mota LM, dos Santos Neto LL, Pereira IA, Burlingame R, Ménard HA,
Laurindo IM. [Autoantibodies as predictors of biological therapy for early
rheumatoid arthritis]. Acta Reumatologica Portuguesa 2010; 35(2):156-166.
[Article in Portuguese].
43. Braun-Moscovici Y, Markovits D, Zinder O, SchapiraD, Rozin A, Ehrenburg M,
Dain L, Hoffer E, Nahir AM, Balbir-Gurman A. Anti-cyclic citrullinated protein
149
antibodies as a predictor of response to anti-tumor necrosis factor-alpha therapy
in patients with rheumatoid arthritis. The Journal of Rheumatology 2006;
33(3):497-500.
44. Katchamart W, Johnson S, Lin HJ, Phumethum V, Salliot C, Bombardier C.
Predictors for remission in rheumatoid arthritis patients: A systemic review.
Arthritis Care and Research (Hoboken) 2010; 62(8):1128-1143.
45. Hyrich KL, Watson KD, Silman AJ, Symmons DP; British Society for
Rheumatology Biologics Register. Predictors of response to anti-TNF-alpha
therapy among patients with rheumatoid arthritis: results from the British
Society for Rheumatology Biologics Register. Rheumatology (Oxford) 2006;
45(12):1558-1565.
46. Bizzaro N, Bartoloni E, Morozzi G, Manganelli S, Riccieri V,
SabatiniP,FilippiniM, Tampoia M, Afeltra A,Sebastiani G, Alpini C, BiniV, Bistoni
O, Alunno A, Gerli R; Forum Interdisciplinare per la Ricerca nelle Malattie
Autoimmuni (FIRMA Group). Anti-cyclic citrullinated peptide antibody titer
predicts time to rheumatoid arthritis onset in patients with undifferentiated
arthritis: results from a 2-year prospective study. Arthritis Research & Therapy
2013; 15(1):R16.
47. Meyer O, Nicaise-Roland P, Santos MD, Labarre C, Dougados M, Goupille P,
Cantagrel A, Sibilia J, Combe B. Serial determination of cyclic citrullinated
peptide autoantibodies predicted five-year radiological outcomes in a
prospective cohort of patients with early rheumatoid arthritis. Arthritis Research
& Therapy 2006; 8(2):R40.
48. Fabre S, Guisset C, Tatem L, Dossat N, Dupuy AM, Cohen JD, Cristol JP,
Daures JP, Jorgensen C. Protein biochip array technology to monitor rituximab
150
in rheumatoid arthritis. Clinical & Experimental Immunology 2009; 155(3):395402.
49. Cuchacovich M, Catalan D, Wainstein E, Gatica H, SotoL, Aravena O, Pesce B,
Sabugo F, Aguillón JC. Basal anti-cyclic citrullinated peptide (anti-CCP)
antibody levels and a decrease in anti-CCP titres are associated with clinical
response to adalimumab in rheumatoid arthritis. Clinical and Experimental
Rheumatology 2008; 26(6):1067-1073.
50. Davis JM 3rd, Matteson EL. [Cytokine biomarkers and the promise of
personalized therapy in rheumatoid arthritis]. Reumatología Clinica 2009;
5(4):143-146. [Article in Spanish].
51. Berglin E, Johansson T, Sundin U, Jidell E, Wadell G, Hallmans G, RantapääDahlqvist S. Radiological outcome in rheumatoid arthritis is predicted by
presence of antibodies against cyclic citrullinated peptide before and at disease
onset, and by IgA-RF at disease onset. Annals of the Rheumatic Diseases
2006; 65(4):453-458.
52. Burr ML, Viatte S, Bukhari M, Plant D, Symmons DP, Thomson W, Barton A.
Long-term stability of anti-cyclic citrullinated peptide antibody status in patients
with early inflammatory polyarthritis. Arthritis Research & Therapy 2012;
14(3):R109.
53. Taylor P, Gartemann J, Hsieh J, Creeden J. A systemic review of serum
biomarkers anti-cyclic citrullinated peptide and rheumatoid factors as tests for
rheumatoid arthritis. Autoimmune Diseases 2011; 2011: Article ID 815038.
54. Miriovsky BJ, Michaud K, Thiele GM, O’Dell JR, Cannon GW, Kerr G, Richards
JS, Johnson D, Caplan L, Reimold A, Hooker R, Mikuls TR. Anti-CCP antibody
and rheumatoid factor concentrations predict greater disease activity in men
151
with rheumatoid arthritis. Annals of the Rheumatic Diseases 2010; 69(7):12921297.
55. McGeough CM, Bjourson AJ. Diagnostic, prognostic and theranostic genetic
biomarkers for rheumatoid arthritis. Journal of Clinical & Cellular Immunology
2012; S6:002. doi: 10.4172/2155-9899.S6-002.
56. Marasovic-Krstulovic D, Martinovic-Kaliterna D, Fabijanic D, Morovic-Vergles J.
Are the anti-cyclic citrullinated peptide antibodies independent predictors of
myocardial involvement in patients with active rheumatoid arthritis?
Rheumatology (Oxford) 2011; 50(8):1505-1512.
57. Giles JT, Malayeri AA, Fernandes V, Post W, Blumenthal RS, Bluemke D,
Vogel-Claussen J, Szklo M, Petri M, Gelber AC, Brumback L, Lima J, Bathon
JM. Left ventricular structure and function in patients with rheumatoid arthritis,
as assessed by cardiac magnetic resonance imaging. Arthritis and Rheumatism
2010; 62(4):940-951.
58. Nowak D, Lewandowicz J, Dbkowska B, Marczak J. Combination of
methotrexate and prednizone decreases circulating concentrations of interleukin
1 beta and Interleukin 6 in patients with rheumatoid arthritis. Poor correlation of
cytokine suppression with clinical improvement. International Journal of
Immunopathology and Pharmacology 1999; 12(1):13-21.
59. Visvanathan S, Marini JC, Smolen JS, Clair EW, Pritchard C, Shergy W,
Pendley C, Baker D, Bala M, Gathany T, Han J, Wagner C. Changes in
biomarkers of inflammation and bone turnover and associations with clinical
efficacy following infliximab plus methotrexate therapy in patients with early
rheumatoid arthritis. The Journal of Rheumatology 2007; 34(7):1465-1474.
152
60. Emery P, Dörner T. Optimising treatment in rheumatoid arthritis: a review of
potential biological markers of response. Annals of the Rheumatic Diseases
2011; 70(12):2063-2070.
61. Shimamoto K, Ito T, Ozaki Y, Amuro H, Tanaka A, Hishizawa T, Son Y, Inaba
M, Nomura S. Serum interleukin 6 before and after therapy with tocilizumab Is a
principal biomarker in patients with rheumatoid arthritis. The Journal of
Rheumatology 2013; 40(7):1074-1081.
62. Fabre S, Dupuy AM, Dossat N, Guisset C, Cohen JD, Cristol JP, Daures JP,
Jorgensen C. Protein biochip array technology for cytokine profiling predicts
etanercept responsiveness in rheumatoid arthritis. Clinical & Experimental
Immunology 2008; 153(2):188-195.
63. Hueber W, Tomooka BH, Batliwalla F, Li W, MonachPA, Tibshirani RJ, Van
Vollenhoven RF, Lampa J, SaitoK, TanakaY, Genovese MC. Blood
autoantibody and cytokine profiles predict response to anti-tumor necrosis factor
therapy in rheumatoid arthritis. Arthritis Research & Therapy 2009 ; 11(3):R76.
64. Potter C, Hyrich KL, Tracey A, Lunt M, Plant D, Symmons DP, Thomson W,
Worthington J, Emery P, Morgan AW, Wilson AG, Isaacs J, Barton A;
BRAGGSS. Association of rheumatoid factor and anti-cyclic citrullinated peptide
positivity, but not carriage of share epitope or PTPN22 susceptibility variants,
with anti-tumour necrosis factor response in rheumatoid arthritis. Annals of the
Rheumatic Diseases 2009; 68(1): 69-74.
65. Tchetverikov I, Lard LR, DeGroot J, Verzijl N, TeKoppele JM, Breedveld FC,
Huizinga TW, Hanemaaijer R. Matrix metalloproteinases-3, -8, -9 as markers of
disease activity and joint damage progression in early rheumatoid arthritis.
Annals of the Rheumatic Diseases 2003; 62(11):1094-1099.
153
66. Green MJ, Gough AK, Devlin J, Smith J, Astin P, Taylor D, Emery P. Serum
MMP-3 and MMP-1 and progression of joint damage in early rheumatoid
arthritis. Rheumatology [Oxford] 2003; 42(1):83-88.
154
Conclusion
Rheumatoid arthritis is a chronic inflammatory arthropathy of unknown aetiology.
Despite the lack of scientific progress in identifying the underlying causative factors,
significant advances have been made in the understanding of pathogenic
mechanisms involved. The molecular and cellular components of this pathologic
immuno-inflammatory response are now well characterized, enabling the
development of novel successful therapeutic interventions.
RA in the developing world has been shown to be associated with as much, if not
more morbidity than in developed countries, and few longitudinal studies in
developing countries, particularly in sub-Saharan Africa have evaluated clinical,
demographic and serological aspects of RA management. Following the interim
analysis of this cohort of DMARD naïve early RA patients, some aspects deserve
emphasis.
RA patients experienced severe functional impairment despite therapy with only a
third of patients achieving an acceptable level of response at one year under routine
care. At the outset patients experienced considerable physical and psycho-social
impairment with over 60% of patients still having significant disease burden after one
year of therapy. A lower level of schooling, unemployment, high disease activity,
nodulosis and radiographic damage were some of the factors associated with poorer
response to therapy. These factors underscore the need for early diagnosis and an
aggressive goal directed therapeutic regimen. Judicious use of newer therapeutic
modalities is likely to be cost-effective when combined with clinical and laboratory
biomarker strategies, which have the potential to facilitate RA management by
identifying poor prognostic factors, thereby potentially guiding choice of therapy.
Genotyping of the patient cohort revealed a high percentage of patients with the
susceptibility HLA DRB1 shared epitope allele similar to many studies in the
developed world, but in contrast to some studies in Africa. Characterisation of
genotypes according to the new du Montcel classification has not been previously
evaluated in black South Africans with RA, and revealed the S2 and S3P high-risk
155
allele as being pre-dominant with under-representation of the S1 allelle. The
presence of the SE was associated with high disease activity, ACPA and cytokine
profiles, but added no additional value relative to measurement of ACPA. Testing for
the SE is thus not recommended in routine care.
The majority of patients was sero-positive with cytokine profiles demonstrating both a
pro-inflammatory and counter regulatory anti-inflammatory cytokine milieu. The
diagnostic sensitivities of both RF (79%) and ACPA (78%) was similar with increased
specificity for RA if both were positive. Baseline ACPA status was not associated with
features of disease severity and testing is recommended only in the situation of RF
negativity and when clinical suspicion of RA is high. Treatment-naïve cytokine profiles
revealed global elevation of cytokines with high inter-cytokine correlations particularly
in the high disease activity groups, with VEGF also correlating with radiologic damage
and auto-antibodies in the HDA group (at baseline). Cytokine profiles in patients with
nodulosis suggest an exaggerated Th1 and macrophage response with elevated proangiogenic factors VEGF and IL-8. Baseline DMARD naïve cytokine levels did not
correlate with clinical measures of disease activity or auto-antibodies, but could shed
light on immune-pathogenic mechanisms or have prognostic value.
Biomarkers of cartilage metabolism such as MMP-3 were associated with disease
activity at baseline and predictive of a higher disease activity at 6 months. Elevated
baseline serum MMP-3 levels were predictive (OR 2.8) of higher disease activity
(SDAI>11) at 6 months follow up, possibly identifying a subset of patients likely to
benefit from more aggressive management. None of the auto-antibodies or cytokine
profiles at baseline were predictive of disease activity at 6 months.
The recommended therapeutic goal is to have no more than a low disease activity
state within 6 months of diagnosis. Characterization of auto-antibody and cytokine
profiles at this stage has the potential to stratify patients and guide future therapies.
Auto-antibody and cytokine levels following synthetic DMARDs decreased
significantly in tandem with disease activity. A more robust decline in cytokines was
seen in patients having achieved low disease activity and in patients treated with a
156
combination of MTX and steroids. Lack of correlation of auto-antibody and cytokine
levels with measures of disease activity preclude the utilty of serial measurement.
Limitations of the study include; i) The relatively small number of patients particularly
when analysing sub-groups; ii) patients managed as per routine care thus treatment
not randomised; iii) patients lost to follow up (20% at 1 year); and iv) full panel of
cytokines not tested at 6 months due to cost constraints.
The strength of the study lies in the prospective review of clinical and serological
measures associated with RA in a ‘real life’ situation, potentially providing practical
cost-effective recommendations and highlighting the need for more research. In a
resource limited country early diagnosis and aggressive management targeting of at
least a low disease activity state within 6 months should be achieved in most of our
patients. Management should include patient education, social support and rapid
escalation of DMARDs. This will require more frequent and intensive review putting
more strain on health providers thus it is imperative that patients with poor prognostic
markers be identified at the outset. Expensive biologic therapies are accessible to a
only few patients thus characterisation of cytokine profile in sDMARD refractory
patients may facilitate proper patient selection and choice of therapy. In addition to
clinical and functional measures, a useful outcome measure, radiographic
progression is also being analysed in this cohort at 1 and 2 years after treatment.
Biomarkers including baseline auto-antibodies, cytokines and cartilage metabolites
able to predict radiographic progression may be used to identify patients requiring a
more extensive yet cost effective- approach.
157
Appendix 1
Abstracts of data published as Co-author
1.1
Meyer PW, Hodkinson B, Ally M, Musenge E, Wadee AA, Fickl H, Tikly M,
Anderson R. Circulating cytokine profiles and their relationships with autoantibodies,
acute phase reactants, and disease activity in patients with rheumatoid arthritis.
Mediators of Inflammation 2010; 2010:158514.
Our objective was to analyse the relationship between circulating cytokines,
autoantibodies, acute phase reactants, and disease activity in DMARDs-naïve
rheumatoid arthritis (RA) patients (n = 140). All cytokines were significantly
higher in the RA cohort than in healthy controls. Moderate-to-strong positive
intercorrelations were observed between Th1/Th2/macrophage/fibroblastderived cytokines. RF correlated significantly with IL-1β, IL-2, IL-4, IL-10, IL12, G-CSF, GM-CSF, IFN-γ, and TNF (P < .0001), and aCCP and aMCV with
IL-1β, IL-2, IL-4, and IL-10 (P < .0002), while IL-6 correlated best with the
acute phase reactants, CRP, and SAA (P < .0001). In patients with a DAS28
score of ≥5.1, IFN-γ, IL-1β, IL-1Ra, TNF, GM-CSF, and VEGF were
significantly correlated (P < .04-.001) with high disease activity (HDA).
Circulating cytokines in RA reflect a multifaceted increase in immune reactivity
encompassing Th1 and Th2 cells, monocytes/macrophages, and synovial
fibroblasts, underscored by strong correlations between these cytokines, as
well as their relationships with RF, aCCP, and aMCV, with some cytokines
showing promise as biomarkers of HDA.
1.2
Hodkinson B, Meyer PW, Musenge E, Ally MM, Wadee AA, Anderson R, Tikly
M. The diagnostic utility of the anti-CCP antibody test is no better than rheumatoid
factor in South Africans with early rheumatoid arthritis. Clin Rheumatol 2010;
29(6):615-618.
158
To establish the diagnostic utility of the anti-cyclic-citrullinated peptide antibody
(aCCP) test in Black South Africans with early rheumatoid arthritis (RA). A
cross-sectional study comparing the rheumatoid factor (RF) and aCCP status
in RA patients and a control group consisting of healthy subjects, and patients
with systemic lupus erythematosus (SLE) and scleroderma. The sensitivity,
specificity, positive (PPV) and negative predictive values of the aCCP test
alone were 82.5%, 84.9%, 87.6% and 79% versus 81.7%, 90.7%, 92.5% and
78% for RF alone. The best specificity (95.3) and PPV (95.8%) was observed
when both aCCP and RF tests were positive. Patients with erosive disease
had a significantly higher mean RF titre compared with those with non-erosive
disease (p = 0.007). There was a trend towards an association of smoking (OR
= 4.1, 95% CI = 0.9-18.6) and functional disability (p = 0.07) with RF-positive
status. No similar clinical associations were observed with aCCP. Almost a
third of SLE patients were aCCP positive. Despite the best specificity and PPV
observed when both the aCCP and RF tests were positive, our findings
suggest that testing for aCCP is only cost-effective in the RF-negative patient
in whom there is a strong clinical suspicion of RA.
1.3
Meyer PW, Hodkinson B, Ally M, Musenge E, Wadee AA, Fickl H, Tikly M,
Anderson R. HLA-DRB1 shared epitope genotyping using the revised classification
and its association with circulating autoantibodies, acute phase reactants, cytokines
and clinical indices of disease activity in a cohort of South African rheumatoid arthritis
patients. Arthritis Res Ther 2011; 13(5):R160.
INTRODUCTION:
The revised shared epitope (SE) concept in rheumatoid arthritis (RA) is based
on the presence (S) or absence (X) of the SE RAA amino acid motif at
positions 72 to 74 of the third hypervariable region of the various human
leucocyte antigen (HLA)-DRB1 alleles. The purpose of this study was to
investigate SE subtypes on the basis of the American College of
Rheumatology 1987 revised criteria for the classification of RA in a cohort of
159
South African RA patients (n = 143) and their association with clinical and
circulating biomarkers of disease activity (autoantibodies, acute phase
reactants and cytokines).
METHODS:
Genomic DNA was analysed using high-resolution recombinant sequencespecific oligonucleotide PCR typing of the HLA-DRB1 allele. Subtypes of the
SE were classified according to the amino acids at positions 72 to 74 for the
RAA sequence, and further sub-divided according to the amino acids at
positions 70 and 71, which either contribute to (S2, S3P), or negate (S1, S3D)
RA susceptibility. Disease activity was assessed on the basis of (1) Disease
Activity Score in 28 joints using C-reactive protein (CRP), (2) rheumatoid factor
(RF), (3) CRP and (4) serum amyloid A by nephelometry, anticyclic
citrullinated peptide antibodies (aCCP) by an immunofluorometric procedure,
and cytokines by multiplex bead array technology.
RESULTS:
Of the 143 RA patients, 81 (57%) were homozygous (SS) and 50 (35%) were
heterozygous (SX) for the SE alleles with significant overexpression of S2 and
S3P (respective odds ratios (ORs) 5.3 and 5.8; P < 0.0001), and 12 (8%) were
classified as no SE allele (XX). Both the SS and SX groups showed a strong
association with aCCP positivity (OR = 10.2 and P = 0.0010, OR = 9.2 and P =
0.0028, respectively) relative to the XX group. Clinical scores and
concentrations of the other biomarkers of disease activity (RF, CRP and T
helper cell type 1 (Th1), Th2, macrophage and fibroblast cytokines) were also
generally higher in the SS group than in the SX and XX groups.
CONCLUSIONS:
RA susceptibility alleles investigated according to revised criteria for the
classification of RA were significantly increased in South African RA patients
160
and strongly associated with aCCP in particular as well as with circulating
cytokines and disease severity.
1.4
Hodkinson B, Musenge E, Ally M, Meyer PW, Anderson R, Tikly M.
Functional disability and health-related quality of life in South Africans with early
rheumatoid arthritis. Scand J Rheumatol. 2012 Oct;41(5):366-74.
BACKGROUND:
The severity and predictors of functional disability and health-related quality of
life (HRQoL) in a cohort of South Africans with early rheumatoid arthritis (RA)
were investigated.
METHODS:
Changes in the Health Assessment Questionnaire Disability Index (HAQ) and
the 36-Item Short Form Health Survey (SF-36) following 12 months of
traditional disease-modifying anti-rheumatic drugs (DMARDs) were studied in
previously DMARD-naïve adults with disease duration ≤ 2 years.
RESULTS:
The majority of the 171 patients were female (82%), Black Africans (89%) with
a mean (SD) symptom duration of 11.6 (7.0) months. In the 134 patients seen
at 12 months, there were significant improvements in the HAQ and all domains
of the SF-36 but 92 (69%) still had substantial functional disability (HAQ > 0.5)
and 89 (66%) had suboptimal mental health [SF-36 mental composite score
(MCS) < 66.6]. Multivariate analysis showed that female sex (p = 0.05) and
high baseline HAQ score (p < 0.01) predicted substantial functional disability at
12 months. Unemployment (p = 0.03), high baseline pain (p = 0.02), and HAQ
score (p = 0.04) predicted suboptimal mental health, with a trend towards a
low level of schooling being significant (p = 0.08).
161
CONCLUSIONS:
Early RA has a broad impact on HRQoL in indigent South Africans, with a
large proportion of patients still showing substantial functional disability and
suboptimal mental health despite 12 months of DMARD therapy. Further
research is needed to establish the role of interventions including psychosocial
support, rehabilitation programmes, and biological therapy to improve physical
function and HRQoL in this population.
1.5
Hodkinson B, Musenge E, Ally M, Meyer PW, Anderson R, Tikly M. Response
to traditional disease-modifying anti-rheumatic drugs in indigent South Africans with
early rheumatoid arthritis. Clin Rheumatol 2012; 31(4):613-9.
The clinical response to traditional disease-modifying anti-rheumatic drugs
(DMARDs) in indigent South Africans with early rheumatoid arthritis was
investigated. A cohort of patients with early (≤2 years) RA who were DMARDnaïve at inception were prospectively assessed for response to DMARDs
using the Simplified Disease Activity Index (SDAI) over a 12-month period.
Patients with low disease activity (LDA) at 12 months were compared to those
with moderate and high disease activity with respect to demographic, clinical,
autoantibody and radiographic features. The 171 patients (140 females) had a
mean (SD) age of 47.1 (12.4) years, symptom duration of 11.7 (7.1) months
and baseline SDAI of 39.4 (16.2). There was a significant overall improvement
in the SDAI and its components in the 134 (78.4%) patients who completed the
12 months visit, but only 28.4% of them achieved LDA. The majority of patients
(91%) were treated with methotrexate as monotherapy or in combination with
chloroquine and/or sulphasalazine. Baseline features that independently
predicted a LDA state at 12 months were lower Health Assessment
Questionnaire Disability Index (p = 0.023) and a higher haemoglobin level (p =
0.048). Receiver operating characteristic curve analysis showed that the 6month SDAI was better than the baseline SDAI in predicting the 12-month
SDAI (area under the curve of 0.69 vs. 0.52, respectively, p = 0.008). In
162
conclusion, less than a third of the patients achieved a low disease activity at
12 months on traditional DMARDs. Patients who have an inadequate response
to traditional DMARDs at 6 months are unlikely to show further improvement
on traditional DMARDs at 12 months. These findings underscore the need for
better disease control by an aggressive tight control strategy, including intense
patient education and biologic therapy.
1.6
Hodkinson B, Meyer PW, Musenge E, Ally M, Anderson R, Tikly
M.Exaggerated circulating Th-1 cytokine response in early rheumatoid arthritis
patients with nodules. Cytokine 2012; 60(2):561-564.
BACKGROUND:
Immunohistochemical studies of the rheumatoid nodule (RN) suggest it is a
Th1 granuloma, with focal vasculitis, yet the pathogenesis remains unclear and
little is known about circulating cytokines in these patients.
OBJECTIVE:
We studied circulating cytokines in DMARD-naïve RA patients to investigate
associations with subcutaneous RN.
METHODS:
149 DMARD-naïve adults with early RA (symptom duration ≤ 2 years) were
assessed using the Simplified Disease Activity Index (SDAI), and hand and
feet radiographs were scored using the modified Larsen method. Circulating
cytokines and growth factors representative of T-helper cell 1(Th1) and Th2
cell, macrophages, and fibroblasts were measured using the Bio-Plex®
suspension array system.
163
RESULTS:
Of 149 patients, 34 (22.8%) had subcutaneous RN, and these patients had
more severe disease with higher mean swollen joint counts (p=0.02), SDAI
(p=0.04) and modified Larsen scores (p=0.004). There were no differences in
Rheumatoid Factor or anti-cyclic citrullinated peptide antibody positivity
between patients with RN and those without RN. Patients with RN showed
significantly higher levels of circulating IL-12 (p=0.02), IL-2 (p=0.048), and
VEGF (p=0.033) levels, with a trend towards higher levels of IL-7 (p=0.056),
IFN-γ (p=0.059) and IL-8 (p=0.074) compared to those without RN.
CONCLUSIONS:
DMARD-naïve early RA patients with RN had more severe disease than those
without RN, and showed an exaggerated circulating Th1 and macrophage
cytokine profile.
164
Appendix 2
Clinical Record
CENTRE NUMBER
PATIENT NUMBER
PATIENT INITIALS
DATE ___/___/___
ASSESSOR INITIALS
EARLY MORNING STIFFNESS (minutes) _________________________
SWOLLEN JOINT COUNT
RIGHT
Shoulder
LEFT
Elbow
Wrist
5
4
3
2
1
MCP
1
2
3
4
5
PIP
Knee
TENDER JOINT COUNT : _________
RIGHT
Shoulder
LEFT
Elbow
Wrist
5
4
3
2
1
MCP
1
2
3
4
5
PIP
Knee
165
PHYSICIAN GLOBAL ASSESSMENT
Excellent
Good
Fair
Poor
Very poor
PATIENT PAIN ASSESSMENT
How bad has your pain over the last week?
No pain
Mild pain
Moderate pain
Severe pain
Unbearable pain
PATIENT GLOBAL ASSESSMENT
How has your arthritis been over last week?
Excellent
Good
Fair
Poor
Very poor
166